1
|
Qi L, Wang J, Yan J, Jiang W, Ge W, Fang X, Wang X, Shen SG, Liu L, Zhang L. Engineered extracellular vesicles with sequential cell recruitment and osteogenic functions to effectively promote senescent bone repair. J Nanobiotechnology 2025; 23:107. [PMID: 39939879 PMCID: PMC11823168 DOI: 10.1186/s12951-025-03168-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/25/2025] [Indexed: 02/14/2025] Open
Abstract
Senescent mandibular bone repair poses a formidable challenge without a completely satisfactory strategy. Endogenous cell recruitment and osteogenic differentiation are two sequential stages in bone regeneration, and disruptions in these two processes present significant obstacles to senescent bone repair. To address these issues, engineered extracellular vesicles (EV) with sequential stem cell recruitment and osteogenic functions were developed. This study demonstrated that Apt19s-engineered extracellular vesicles (Apt19s-EV) recognize and recruit bone marrow mesenchymal stem cells derived from old rats (O-BMSCs) specifically and effectively. MiR-376b-5p, identified by RNA sequencing and transfection, was significantly decreased in O-BMSCs, and it was selected to construct miR-376b-5p-engineered extracellular vesicles (376b-EV). 376b-EV could promote osteogenesis and alleviate senescence of O-BMSCs by targeting Camsap1. To combine the advantages of Apt19s and miR-376b-5p, dual engineered extracellular vesicles (Apt-376b-EV) comprising both Apt19s and miR-376b-5p modifications were constructed. To further validate its function, Gelatin methacryloyl (GelMA) hydrogel was used as a carrier to construct the Apt-376b-EV@GelMA delivery system. The in vitro results have demonstrated that Apt-376b-EV@GelMA could recruit O-BMSCs, alleviate senescence and promote osteogenic differentiation sequentially. Notably, the in vivo study also showed that Apt-376b-EV@GelMA could sequentially recruit endogenous stem cells and enhance new bone formation in senescent bone fracture and critical-sized defect models. In summary, the dual engineered extracellular vesicles, Apt-376b-EV, offer an appealing solution for recruiting endogenous stem cells and promoting bone repair sequentially in the senescent microenvironment, which may broaden the clinical applications of engineered EV and provide valuable strategies for treating senescent bone-related diseases in the future clinical work.
Collapse
Affiliation(s)
- Lei Qi
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China
| | - Jing Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China
| | - Jinge Yan
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China
| | - Weidong Jiang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China
| | - Weiwen Ge
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China
| | - Xin Fang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China
| | - Xudong Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China
| | - Steve Gf Shen
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China.
| | - Lu Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China.
| | - Lei Zhang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai, 200011, PR China.
| |
Collapse
|
2
|
Müller D, Klotsche J, Kosik MB, Perka C, Buttgereit F, Hoff P, Gaber T. Fracture Fusion on Fast-Forward: Locally Administered Deferoxamine Significantly Enhances Fracture Healing in Animal Models: A Systematic Review and Meta-Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413290. [PMID: 39840407 PMCID: PMC11848589 DOI: 10.1002/advs.202413290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/08/2024] [Indexed: 01/23/2025]
Abstract
Fractures, with a yearly incidence of 1.2%, can lead to healing complications in up to 10% of cases. The angiogenic stimulant deferoxamine (DFO) is recognized for enhancing bone healing when administered into the fracture gap. This systematic review with meta-analysis investigates the effect of local DFO application on bone healing in rat and mouse models. EMBASE, MEDLINE (PubMed), and Web of Science are systematically searched in January 2024. The study is prospectively registered in PROSPERO (CRD42024492533), and the SYRCLE tool is used to assess study quality and risk of bias. Outcome values contain the primary endpoint bone volume fraction (BV/TV) as well as the secondary endpoints bone volume, tissue volume, bone mineral density, trabecular separation, trabecular thickness, vessel formation and the mechanical properties, assessed by µCT, angiography and mechanical strength tests. Out of 21 included studies, 18 qualify for meta-analysis, involving 539 animals. DFO-treated groups exhibit significantly higher BV/TV values (p < 0.0001) compared to controls, with similarly significant improvements in secondary outcomes. These findings highlight the substantial benefit of DFO in promoting bone healing, especially after radiotherapy. Rapid clinical implementation is recommended to help patients at high risk of fracture healing complications.
Collapse
Affiliation(s)
- Daniel Müller
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
| | - Jens Klotsche
- Deutsches Rheumaforschungszentrum Berlin (DRFZ)a Leibniz Institute10117BerlinGermany
| | - Magdalena B. Kosik
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
| | - Carsten Perka
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinCharitéCenter for Orthopedics und Traumatology10117BerlinGermany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ)a Leibniz Institute10117BerlinGermany
| | - Paula Hoff
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
- MVZ Endokrinologikum Berlin am Gendarmenmarkt10117BerlinGermany
| | - Timo Gaber
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ)a Leibniz Institute10117BerlinGermany
| |
Collapse
|
3
|
Synthesis of novel benzothiophene derivatives as protectors against cranial irradiation-induced neuroinflammation. Future Med Chem 2022; 14:1527-1539. [DOI: 10.4155/fmc-2022-0203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Cranial irradiation results in many deleterious effects to normal tissues, including neuroinflammation. There is a need to explore radioprotective agents that could be safely used to ameliorate these effects. Method: Nine novel benzothiophene derivatives bearing pyrimidinone, pyrazolidinone, triazole and other active moieties were synthesized and evaluated as antioxidants in an in vitro screening experiment. The most potent compounds were then tested as protectors against radiation-induced neuroinflammation and oxidative stress in rat brains following cranial irradiation. Results: The most potent antioxidant compounds were compounds 3–5 and 10 . P-fluro,p- bromo and pyrido benzothiophene derivatives offered good antioxidant and anti-inflammatory effects. Conclusion: Compounds 3–5 may be introduced as nontoxic candidates for adjuvant therapeutic protocols used in head and neck tumor radiotherapeutic management.
Collapse
|
4
|
Wei F, Neal CJ, Sakthivel TS, Fu Y, Omer M, Adhikary A, Ward S, Ta KM, Moxon S, Molinari M, Asiatico J, Kinzel M, Yarmolenko SN, San Cheong V, Orlovskaya N, Ghosh R, Seal S, Coathup M. A novel approach for the prevention of ionizing radiation-induced bone loss using a designer multifunctional cerium oxide nanozyme. Bioact Mater 2022; 21:547-565. [PMID: 36185749 PMCID: PMC9507991 DOI: 10.1016/j.bioactmat.2022.09.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
The disability, mortality and costs due to ionizing radiation (IR)-induced osteoporotic bone fractures are substantial and no effective therapy exists. Ionizing radiation increases cellular oxidative damage, causing an imbalance in bone turnover that is primarily driven via heightened activity of the bone-resorbing osteoclast. We demonstrate that rats exposed to sublethal levels of IR develop fragile, osteoporotic bone. At reactive surface sites, cerium ions have the ability to easily undergo redox cycling: drastically adjusting their electronic configurations and versatile catalytic activities. These properties make cerium oxide nanomaterials fascinating. We show that an engineered artificial nanozyme composed of cerium oxide, and designed to possess a higher fraction of trivalent (Ce3+) surface sites, mitigates the IR-induced loss in bone area, bone architecture, and strength. These investigations also demonstrate that our nanozyme furnishes several mechanistic avenues of protection and selectively targets highly damaging reactive oxygen species, protecting the rats against IR-induced DNA damage, cellular senescence, and elevated osteoclastic activity in vitro and in vivo. Further, we reveal that our nanozyme is a previously unreported key regulator of osteoclast formation derived from macrophages while also directly targeting bone progenitor cells, favoring new bone formation despite its exposure to harmful levels of IR in vitro. These findings open a new approach for the specific prevention of IR-induced bone loss using synthesis-mediated designer multifunctional nanomaterials.
Collapse
Key Words
- ALP, Alkaline phosphatase
- BMSC, Bone marrow derived mesenchymal stem cells
- Bone resorption
- Bone strength
- CAT, Catalase
- COLI, Collagen type I
- CTSK, Cathepsin K
- CTX-1, Cross-linked C-telopeptide of type I collagen
- CeONPs, Cerium oxide nanoparticles
- Cerium oxide
- DFT, Density functional theory
- DNA, Deoxyribonucleic acid
- EPR, Electron paramagnetic resonance
- FDA, Food and Drug Administration
- GPX, Glutathione peroxidase
- Gy, Gray
- HIF1α, Hypoxia-inducible factor 1 alpha
- IL-1β, Interleukin 1 beta
- IL-6, Interleukin 6
- IR, Ionizing radiation
- Ionizing radiation
- MNGC, Multinucleated giant cell
- Nanozyme
- OCN, Osteocalcin
- Osteoporosis
- RANKL, Receptor activator of nuclear factor kappa-Β ligand
- ROS, Reactive oxygen species
- SAED, Selected area electron diffraction
- SOD, Superoxide dismutase
- TRAP, Tartrate-resistant acid phosphatase
- XPS, X-ray photoelectron spectroscopy
Collapse
Affiliation(s)
- Fei Wei
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Craig J. Neal
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | | | - Yifei Fu
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Mahmoud Omer
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, MI, MI, USA
| | - Samuel Ward
- Department of Chemistry, Oakland University, Rochester, MI, MI, USA
| | - Khoa Minh Ta
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Samuel Moxon
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Marco Molinari
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Jackson Asiatico
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Michael Kinzel
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Sergey N. Yarmolenko
- Engineering Research Center for Revolutionizing Biomaterials, North Carolina A & T University, Greensboro, NC, USA
| | - Vee San Cheong
- Department of Automatic Control and Systems Engineering, Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield, S1 3JD, UK
| | - Nina Orlovskaya
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Ranajay Ghosh
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Melanie Coathup
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
- Corresponding author. Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
5
|
Optimization of a Tricalcium Phosphate-Based Bone Model Using Cell-Sheet Technology to Simulate Bone Disorders. Processes (Basel) 2022. [DOI: 10.3390/pr10030550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Bone diseases such as osteoporosis, delayed or impaired bone healing, and osteoarthritis still represent a social, financial, and personal burden for affected patients and society. Fully humanized in vitro 3D models of cancellous bone tissue are needed to develop new treatment strategies and meet patient-specific needs. Here, we demonstrate a successful cell-sheet-based process for optimized mesenchymal stromal cell (MSC) seeding on a β-tricalcium phosphate (TCP) scaffold to generate 3D models of cancellous bone tissue. Therefore, we seeded MSCs onto the β-TCP scaffold, induced osteogenic differentiation, and wrapped a single osteogenically induced MSC sheet around the pre-seeded scaffold. Comparing the wrapped with an unwrapped scaffold, we did not detect any differences in cell viability and structural integrity but a higher cell seeding rate with osteoid-like granular structures, an indicator of enhanced calcification. Finally, gene expression analysis showed a reduction in chondrogenic and adipogenic markers, but an increase in osteogenic markers in MSCs seeded on wrapped scaffolds. We conclude from these data that additional wrapping of pre-seeded scaffolds will provide a local niche that enhances osteogenic differentiation while repressing chondrogenic and adipogenic differentiation. This approach will eventually lead to optimized preclinical in vitro 3D models of cancellous bone tissue to develop new treatment strategies.
Collapse
|
6
|
Lang A, Stefanowski J, Pfeiffenberger M, Wolter A, Damerau A, Hemmati-Sadeghi S, Haag R, Hauser AE, Löhning M, Duda GN, Hoff P, Schmidt-Bleek K, Gaber T, Buttgereit F. MIF does only marginally enhance the pro-regenerative capacities of DFO in a mouse-osteotomy-model of compromised bone healing conditions. Bone 2022; 154:116247. [PMID: 34743042 DOI: 10.1016/j.bone.2021.116247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022]
Abstract
The initial phase of fracture healing is crucial for the success of bone regeneration and is characterized by an inflammatory milieu and low oxygen tension (hypoxia). Negative interference with or prolongation of this fine-tuned initiation phase will ultimately lead to a delayed or incomplete healing such as non-unions which then requires an effective and gentle therapeutic intervention. Common reasons include a dysregulated immune response, immunosuppression or a failure in cellular adaptation to the inflammatory hypoxic milieu of the fracture gap and a reduction in vascularizing capacity by environmental noxious agents (e.g. rheumatoid arthritis or smoking). The hypoxia-inducible factor (HIF)-1α is responsible for the cellular adaptation to hypoxia, activating angiogenesis and supporting cell attraction and migration to the fracture gap. Here, we hypothesized that stabilizing HIF-1α could be a cost-effective and low-risk prevention strategy for fracture healing disorders. Therefore, we combined a well-known HIF-stabilizer - deferoxamine (DFO) - and a less known HIF-enhancer - macrophage migration inhibitory factor (MIF) - to synergistically induce improved fracture healing. Stabilization of HIF-1α enhanced calcification and osteogenic differentiation of MSCs in vitro. In vivo, only the application of DFO without MIF during the initial healing phase increased callus mineralization and vessel formation in a preclinical mouse-osteotomy-model modified to display a compromised healing. Although we did not find a synergistically effect of MIF when added to DFO, our findings provide additional support for a preventive strategy towards bone healing disorders in patients with a higher risk by accelerating fracture healing using DFO to stabilize HIF-1α.
Collapse
Affiliation(s)
- Annemarie Lang
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany
| | - Jonathan Stefanowski
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Moritz Pfeiffenberger
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Angelique Wolter
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Alexandra Damerau
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Shabnam Hemmati-Sadeghi
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Anja E Hauser
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Max Löhning
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Julius Wolff Institute, Berlin, Germany
| | - Paula Hoff
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Julius Wolff Institute, Berlin, Germany
| | - Timo Gaber
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany.
| | - Frank Buttgereit
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
7
|
Overcoming Nuclear Winter: The Cutting-edge Science of Bone Healing and Regeneration in Irradiated Fields. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2021; 9:e3605. [PMID: 34235033 PMCID: PMC8245112 DOI: 10.1097/gox.0000000000003605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/08/2021] [Indexed: 11/25/2022]
Abstract
Background: The incidence of cancer worldwide is expected to be more than 22 million annually by 2030. Approximately half of these patients will likely require radiation therapy. Although radiotherapy has been shown to improve disease control and increase survivorship, it also results in damage to adjacent healthy tissues, including the bone, which can lead to devastating skeletal complications, such as nonunion, pathologic fractures, and osteoradionecrosis. Pathologic fractures and osteoradionecrosis are ominous complications that can result in large bone and soft tissue defects requiring complex reconstruction. Current clinical management strategies for these conditions are suboptimal and dubious at best. The gold standard in treatment of severe radiation injury is free tissue transfer; however, this requires a large operation that is limited to select candidates. Methods: With the goal to expand current treatment options and to assuage the devastating sequelae of radiation injury on surrounding normal tissue, our laboratory has performed years of translational studies aimed at remediating bone healing and regeneration in irradiated fields. Three therapeutics (amifostine, deferoxamine, and adipose-derived stem cells) have demonstrated great promise in promoting healing and regeneration of irradiated bone. Results: Amifostine confers prophylactic protection, whereas deferoxamine and adipose-derived stem cells function to remediate postradiation associated injury. Conclusions: These prospective therapeutics exploit a mechanism attributed to increasing angiogenesis and ultimately function to protect or restore cellularity, normal cellular function, osteogenesis, and bone healing to nonirradiated metrics. These discoveries may offer innovative treatment alternatives to free tissue transfer with the added benefit of potentially preventing and treating osteoradionecrosis and pathologic fractures
Collapse
|
8
|
Katsila T, Chasapi SA, Gomez Tamayo JC, Chalikiopoulou C, Siapi E, Moros G, Zoumpoulakis P, Spyroulias GA, Kardamakis D. Three-Dimensional Cell Metabolomics Deciphers the Anti-Angiogenic Properties of the Radioprotectant Amifostine. Cancers (Basel) 2021; 13:cancers13122877. [PMID: 34207535 PMCID: PMC8230228 DOI: 10.3390/cancers13122877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cancer and inflammation share aberrant angiogenesis as a hallmark, and, thus, anti-angiogenetic strategies remain of key interest. Amifostine, which is already a drug on the market, may be of further benefit to patients also in the context of drug repurposing. To shed light on the anti-angiogenic properties of amifostine during human adult angiogenesis and grasp the early events of angiogenesis, we employed 3D cell untargeted metabolomics by liquid chromatography–mass spectrometry and nuclear magnetic resonance spectroscopy in the presence of vascular endothelial growth factor-A or deferoxamine (pro-angiogenic factors that exhibit distinct angiogenesis induction profiles). Our findings reveal mechanism-specific inhibitory profiles of amifostine against VEGF-A- and deferoxamine-induced angiogenesis. Amifostine may serve as a dual radioprotective and anti-angiogenic agent in radiotherapy patients. Abstract Aberrant angiogenesis is a hallmark for cancer and inflammation, a key notion in drug repurposing efforts. To delineate the anti-angiogenic properties of amifostine in a human adult angiogenesis model via 3D cell metabolomics and upon a stimulant-specific manner, a 3D cellular angiogenesis assay that recapitulates cell physiology and drug action was coupled to untargeted metabolomics by liquid chromatography–mass spectrometry and nuclear magnetic resonance spectroscopy. The early events of angiogenesis upon its most prominent stimulants (vascular endothelial growth factor-A or deferoxamine) were addressed by cell sprouting measurements. Data analyses consisted of a series of supervised and unsupervised methods as well as univariate and multivariate approaches to shed light on mechanism-specific inhibitory profiles. The 3D untargeted cell metabolomes were found to grasp the early events of angiogenesis. Evident of an initial and sharp response, the metabolites identified primarily span amino acids, sphingolipids, and nucleotides. Profiles were pathway or stimulant specific. The amifostine inhibition profile was rather similar to that of sunitinib, yet distinct, considering that the latter is a kinase inhibitor. Amifostine inhibited both. The 3D cell metabolomics shed light on the anti-angiogenic effects of amifostine against VEGF-A- and deferoxamine-induced angiogenesis. Amifostine may serve as a dual radioprotective and anti-angiogenic agent in radiotherapy patients.
Collapse
Affiliation(s)
- Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (C.C.); (E.S.); (G.M.); (P.Z.)
- Department of Radiation Oncology, University of Patras Medical School, 26504 Patras, Greece;
- Correspondence: ; Tel.: +30-210-727-3752
| | - Styliani A. Chasapi
- Department of Pharmacy, University of Patras, 26504 Patras, Greece; (S.A.C.); (G.A.S.)
| | | | - Constantina Chalikiopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (C.C.); (E.S.); (G.M.); (P.Z.)
| | - Eleni Siapi
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (C.C.); (E.S.); (G.M.); (P.Z.)
| | - Giorgos Moros
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (C.C.); (E.S.); (G.M.); (P.Z.)
| | - Panagiotis Zoumpoulakis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (C.C.); (E.S.); (G.M.); (P.Z.)
| | | | - Dimitrios Kardamakis
- Department of Radiation Oncology, University of Patras Medical School, 26504 Patras, Greece;
| |
Collapse
|
9
|
Angiogenesis in Wound Healing following Pharmacological and Toxicological Exposures. CURRENT PATHOBIOLOGY REPORTS 2020. [DOI: 10.1007/s40139-020-00212-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
10
|
Dréno M, Bléry P, Guicheux J, Weiss P, Malard O, Espitalier F. Development of a Rat Model of Mandibular Irradiation Sequelae for Preclinical Studies of Bone Repair. Tissue Eng Part C Methods 2020; 26:447-455. [PMID: 32729379 DOI: 10.1089/ten.tec.2020.0109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Repairing mandibular bone defects after radiotherapy of the upper aerodigestive tract is clinically challenging. Although bone tissue engineering has recently generated a number of innovative treatment approaches for osteoradionecrosis (ORN), these modalities must be evaluated preclinically in a relevant, reproducible, animal model. The objective of this study was to evaluate a novel rat model of mandibular irradiation sequelae, with a focus on the adverse effects of radiotherapy on bone structure, intraosseous vascularization, and bone regeneration. Rats were irradiated with a single 80 Gy dose to the jaws. Three weeks after irradiation, mandibular bone defects of different sizes (0, 1, 3, or 5 mm) were produced in each hemimandible. Five weeks after the surgical procedure, the animals were euthanized. Explanted mandibular samples were qualitatively and quantitatively assessed for bone formation, bone structure, and intraosseous vascular volume by using micro-computed tomography, scanning electron microscopy, and histology. Twenty irradiated hemimandibles and 20 nonirradiated hemimandibles were included in the study. The bone and vessel volumes were significantly lower in the irradiated group. The extent of bone remodeling was inversely related to the defect size. In the irradiated group, scanning electron microscopy revealed a large number of polycyclic gaps consistent with periosteocytic lysis (described as being pathognomonic for ORN). This feature was correlated with elevated osteoclastic activity in a histological assessment. In the irradiated areas, the critical-sized defect was 3 mm. Hence, our rat model of mandibular irradiation sequelae showed hypovascularization and osteopenia. Impact statement Repairing mandibular bone defects after radiotherapy of the upper aerodigestive tract is clinically challenging. Novel tissue engineering approaches for healing irradiated bone must first be assessed in animal models. The current rat model of mandibular irradiation sequelae is based on tooth extraction after radiotherapy. However, the mucosal sequelae of radiotherapy often prevent the retention of tissue-engineered biomaterials within the bone defect. We used a submandibular approach to create a new rat model of mandibular irradiation sequelae, which enables the stable retention of biomaterials within the bone defect and should thus facilitate the assessment of bone regeneration.
Collapse
Affiliation(s)
- Marine Dréno
- Service d'ORL et de Chirurgie Cervico-Faciale, Centre Hospitalier Universitaire de Nantes, Nantes, France.,INSERM U1229 RMeS, Université de Nantes, Nantes, France.,PHU4 OTONN, Centre Hospitalier Universitaire de Nantes, Hôtel Dieu, Nantes, France
| | - Pauline Bléry
- INSERM U1229 RMeS, Université de Nantes, Nantes, France.,PHU4 OTONN, Centre Hospitalier Universitaire de Nantes, Hôtel Dieu, Nantes, France.,Service d'Odontologie Restauratrice et Chirurgicale, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Jérôme Guicheux
- INSERM U1229 RMeS, Université de Nantes, Nantes, France.,PHU4 OTONN, Centre Hospitalier Universitaire de Nantes, Hôtel Dieu, Nantes, France
| | - Pierre Weiss
- INSERM U1229 RMeS, Université de Nantes, Nantes, France.,PHU4 OTONN, Centre Hospitalier Universitaire de Nantes, Hôtel Dieu, Nantes, France.,Service d'Odontologie Restauratrice et Chirurgicale, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Olivier Malard
- Service d'ORL et de Chirurgie Cervico-Faciale, Centre Hospitalier Universitaire de Nantes, Nantes, France.,INSERM U1229 RMeS, Université de Nantes, Nantes, France.,PHU4 OTONN, Centre Hospitalier Universitaire de Nantes, Hôtel Dieu, Nantes, France
| | - Florent Espitalier
- Service d'ORL et de Chirurgie Cervico-Faciale, Centre Hospitalier Universitaire de Nantes, Nantes, France.,INSERM U1229 RMeS, Université de Nantes, Nantes, France.,PHU4 OTONN, Centre Hospitalier Universitaire de Nantes, Hôtel Dieu, Nantes, France
| |
Collapse
|
11
|
Therapeutic Interventions to Reduce Radiation Induced Dermal Injury in a Murine Model of Tissue Expander Based Breast Reconstruction. Ann Plast Surg 2020; 85:546-552. [PMID: 32187064 DOI: 10.1097/sap.0000000000002264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Radiation therapy (XRT) induced dermal injury disrupts type I collagen architecture. This impairs cutaneous viscoelasticity, which may contribute to the high rate of complications in expander-based breast reconstruction with adjuvant XRT. The objective of this study was to further elucidate the mechanism of radiation-induced dermal injury and to determine if amifostine (AMF) or deferoxamine (DFO) mitigates type I collagen injury in an irradiated murine model of expander-based breast reconstruction. METHODS Female Lewis rats (n = 20) were grouped: expander (control), expander-XRT (XRT), expander-XRT-AMF (AMF), and expander-XRT-DFO (DFO). Expanders were surgically placed. All XRT groups received 28 Gy of XRT. The AMF group received AMF 30 minutes before XRT, and the DFO group used a patch for delivery 5 days post-XRT. After a 20-day recovery period, skin was harvested. Atomic force microscopy and Raman spectroscopy were performed to evaluate type I collagen sheet organization and tissue compositional properties, respectively. RESULTS Type I collagen fibril disorganization was significantly increased in the XRT group compared with the control (83.8% vs 22.4%; P = 0.001). Collagen/matrix ratios were greatly reduced in the XRT group compared with the control group (0.49 ± 0.09 vs 0.66 ± 0.09; P = 0.017). Prophylactic AMF demonstrated a marked reduction in type I collagen fibril disorganization on atomic force microscopy (15.9% vs 83.8%; P = 0.001). In fact, AMF normalized type I collagen organization in irradiated tissues to the level of the nonirradiated control (P = 0.122). Based on Raman spectroscopy, both AMF and DFO demonstrated significant differential protective effects on expanded-irradiated tissues. Collagen/matrix ratios were significantly preserved in the AMF group compared with the XRT group (0.49 ± 0.09 vs 0.69 ± 0.10; P = 0.010). β-Sheet/α-helix ratios were significantly increased in the DFO group compared with the XRT group (1.76 ± 0.03 vs 1.86 ± 0.06; P = 0.038). CONCLUSIONS Amifostine resulted in a significant improvement in type I collagen fibril organization and collagen synthesis, whereas DFO mitigated abnormal changes in collagen secondary structure in an irradiated murine model of expander-based breast reconstruction. These therapeutics offer the ability to retain the native microarchitecture of type I collagen after radiation. Amifostine and DFO may offer clinical utility to reduce radiation induced dermal injury, potentially decreasing the high complication rate of expander-based breast reconstruction with adjuvant XRT and improving surgical outcomes.
Collapse
|
12
|
Zhang L, Huang B, Tang H, Ye X, Yao Y, Gong P, Tang H. Amifostine inhibited the differentiation of RAW264.7 cells into osteoclasts by reducing the production of ROS under 2 Gy radiation. J Cell Biochem 2019; 121:497-507. [PMID: 31267572 DOI: 10.1002/jcb.29247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 06/11/2019] [Indexed: 02/05/2023]
Abstract
Patients with malignant tumors receive radiotherapy, and radiation could harm the skeletal system, leading to radiation-induced osteoporosis. A major cause of this phenomenon is the activation of osteoclasts by radiotherapy. In this study, we studied whether amifostine (AMI) could affect the differentiation of osteoclast precursor cells (RAW264.7 cells) into osteoclasts under 2 gray (Gy) radiation. Four groups were used in the experiment: (a) 0 Gy (no radiation); (b) 0 Gy + AMI; (c) 2 Gy radiation; and (d) 2 Gy radiation + AMI. After radiation, a proliferation assay, a reactive oxygen species (ROS) assay, a comet assay, Trap staining, reverse transcription polymerase chain reaction, and an animal study to test the effect of AMI on osteoclast precursor cells under 2 Gy radiation were conducted. Cell proliferation was significantly inhibited by AMI (P < .05). In addition, 2 Gy radiation led to longer "comet tails", high level of ROS, and more Trap-positive cells in vivo and in vitro (P < .05). Radiation improved the expression of CSTK, NFAT, and Rankl/OPG gene (P < .05), as well as Trap-5b levels in the serum, and decreased bone mineral density. AMI inhibited the differentiation of RAW264.7 cells, shortened the tail moment length of comets, and decreased the level of ROS induced by radiation. The expression of NFAT, CTSK, and Rankl/OPG was decreased by AMI at the detection time point in radiation groups (P < .05). AMI inhibits the maturation and differentiation of osteoclasts under radiation conditions by reducing DNA damage and ROS induced by radiation, thereby reducing the adverse effects of radiation in the skeletal system, indicating that AMI might be used to treat osteoradionecrosis.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China.,Department of Oral Implantology, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Bo Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China.,Department of Oral Implantology, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Haiyang Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China.,Department of Oral Implantology, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Xingchen Ye
- Department of Oral Implantology, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Yang Yao
- Department of Oral Implantology, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Ping Gong
- Department of Oral Implantology, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Hua Tang
- Department of Oral Implantology, West China School of Stomatology, Sichuan University, Chengdu, PR China
| |
Collapse
|
13
|
Amifostine Suppresses the Side Effects of Radiation on BMSCs by Promoting Cell Proliferation and Reducing ROS Production. Stem Cells Int 2019; 2019:8749090. [PMID: 30728842 PMCID: PMC6343176 DOI: 10.1155/2019/8749090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/02/2018] [Accepted: 10/21/2018] [Indexed: 02/07/2023] Open
Abstract
This study is aimed at investigating the effect of amifostine (AMI) on rat bone marrow stromal stem cells (BMSCs) exposed to 2 Gy radiation. The BMSCs were divided into four groups, namely, group A that received 0 Gy radiation, group B that received 0 Gy radiation and AMI, group C that received 2 Gy radiation, and group D that received 2 Gy radiation and AMI. The proliferation, apoptosis, and distribution of BMSCs in the cell cycle, along with their osteogenesis ability, adipogenesis ability, and ROS production, were subsequently examined. The levels of ALP, PPARγ, P53, and TNFα were determined by Western blotting. The results demonstrated that the proliferation of BMSCs and the levels of ALP in group C were much lower than those in group A. The production of ROS and levels of PPARγ, P53, and TNFα in the group that received 2 Gy radiation were much higher than those in group A. Furthermore, the production of ROS and the levels of PPARγ, P53, and TNFα were much lower in group D than in group C. Additionally, the levels of ALP and extent of cell proliferation were much higher in group D than in group C. The results demonstrated the potential of AMI in reducing the side effects of radiation in BMSCs and in treatment of bone diseases caused by radiation.
Collapse
|
14
|
Yan Y, Chen H, Zhang H, Guo C, Yang K, Chen K, Cheng R, Qian N, Sandler N, Zhang YS, Shen H, Qi J, Cui W, Deng L. Vascularized 3D printed scaffolds for promoting bone regeneration. Biomaterials 2019; 190-191:97-110. [DOI: 10.1016/j.biomaterials.2018.10.033] [Citation(s) in RCA: 282] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 10/28/2022]
|
15
|
Zhang J, Qiu X, Xi K, Hu W, Pei H, Nie J, Wang Z, Ding J, Shang P, Li B, Zhou G. Therapeutic ionizing radiation induced bone loss: a review of in vivo and in vitro findings. Connect Tissue Res 2018; 59:509-522. [PMID: 29448860 DOI: 10.1080/03008207.2018.1439482] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Radiation therapy is one of the routine treatment modalities for cancer patients. Ionizing radiation (IR) can induce bone loss, and consequently increases the risk of fractures with delayed and nonunion of the bone in the cancer patients who receive radiotherapy. The orchestrated bone remodeling can be disrupted due to the affected behaviors of bone cells, including bone mesenchymal stem cells (BMSCs), osteoblasts and osteoclasts. BMSCs and osteoblasts are relatively radioresistant compared with osteoclasts and its progenitors. Owing to different radiosensitivities of bone cells, unbalanced bone remodeling caused by IR is closely associated with the dose absorbed. For doses less than 2 Gy, osteoclastogenesis and adipogenesis by BMSCs are enhanced, while there are limited effects on osteoblasts. High doses (>10 Gy) induce disrupted architecture of bone, which is usually related to decreased osteogenic potential. In this review, studies elucidating the biological effects of IR on bone cells (BMSCs, osteoblasts and osteoclasts) are summarized. Several potential preventions and therapies are also proposed.
Collapse
Affiliation(s)
- Jian Zhang
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Xinyu Qiu
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Kedi Xi
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Wentao Hu
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Hailong Pei
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Jing Nie
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Ziyang Wang
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Jiahan Ding
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Peng Shang
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China.,c Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences , Northwestern Polytechnical University , Xi'an , China.,d Research & Development Institute in Shenzhen , Northwestern Polytechnical University, Fictitious College Garden , Shenzhen , China
| | - Bingyan Li
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China
| | - Guangming Zhou
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| |
Collapse
|
16
|
Abstract
OBJECTIVES To determine whether a substantially lowered radiation protocol would provide satisfactory information for the surgeon, using the distal tibia as a model. METHODS Eleven adult cadaveric distal tibia specimens were used to create Orthopaedic Trauma Association (OTA/AO) 43C distal tibia fractures with varying displacements in 2 planes. Each specimen was scanned at 3 modified protocols, which were then subsequently read by both qualified attending orthopaedists and midlevel residents. Observer reliability was evaluated, as well as confidence levels of identifying fracture pattern and treatment protocols. RESULTS On average, there was less than a millimeter of variability in the measured gap to true gap as a whole (mean = 0.74 mm, P < 0.0001). With regard to measurements in gap, pattern, and treatment plans, no significant difference was found between CT images acquired with standard (110 mAs) compared with medium (60 mAs; mean 0.0 mm, P = 1.0; k = 0.14, P = 0.56; k = 0.38, P = 0.13, respectively) and low protocols (45 mAs; mean 0.01 mm, P = 0.95; k = 0.24, P = 0.32; k = 0.31, P = 0.13, respectively). Furthermore, no significant difference was found in measuring step-off across standard, medium, and low radiation dose (0.21 mm, P = 0.46; 0.28 mm, P = 0.39; -0.16 mm, P = 0.48, respectively). CONCLUSION The results of this study show no significant difference when evaluating current standard and low-dose CT scans using less than one-half the amount of exposure. This suggests that in complex extremity fractures, a new CT protocol may potentially be used. Our initial data show promise that we may retain satisfactory imaging to formulate a treatment plan while also reducing the collective radiation burden to the population.
Collapse
|