1
|
Shah SA, Sohail M, Nakielski P, Rinoldi C, Zargarian SS, Kosik-Kozioł A, Ziai Y, Haghighat Bayan MA, Zakrzewska A, Rybak D, Bartolewska M, Pierini F. Integrating Micro- and Nanostructured Platforms and Biological Drugs to Enhance Biomaterial-Based Bone Regeneration Strategies. Biomacromolecules 2025; 26:140-162. [PMID: 39621708 PMCID: PMC11733931 DOI: 10.1021/acs.biomac.4c01133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 01/14/2025]
Abstract
Bone defects resulting from congenital anomalies and trauma pose significant clinical challenges for orthopedics surgeries, where bone tissue engineering (BTE) aims to address these challenges by repairing defects that fail to heal spontaneously. Despite numerous advances, BTE still faces several challenges, i.e., difficulties in detecting and tracking implanted cells, high costs, and regulatory approval hurdles. Biomaterials promise to revolutionize bone grafting procedures, heralding a new era of regenerative medicine and advancing patient outcomes worldwide. Specifically, novel bioactive biomaterials have been developed that promote cell adhesion, proliferation, and differentiation and have osteoconductive and osteoinductive characteristics, stimulating tissue regeneration and repair, particularly in complex skeletal defects caused by trauma, degeneration, and neoplasia. A wide array of biological therapeutics for bone regeneration have emerged, drawing from the diverse spectrum of gene therapy, immune cell interactions, and RNA molecules. This review will provide insights into the current state and potential of future strategies for bone regeneration.
Collapse
Affiliation(s)
- Syed Ahmed Shah
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
- Faculty
of Pharmacy, The Superior University, Lahore 54000, Punjab, Pakistan
| | - Muhammad Sohail
- Faculty
of Pharmacy, Cyprus International University, Nicosia 99258, North Cyprus
| | - Paweł Nakielski
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Chiara Rinoldi
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Alicja Kosik-Kozioł
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Yasamin Ziai
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Mohammad Ali Haghighat Bayan
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Anna Zakrzewska
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Daniel Rybak
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Magdalena Bartolewska
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| |
Collapse
|
2
|
Carney TE, Biggs AE, Miller MA, Mann KA, Oest ME. Therapeutic radiation directly alters bone fatigue strength and microdamage accumulation. J Mech Behav Biomed Mater 2024; 160:106766. [PMID: 39378671 DOI: 10.1016/j.jmbbm.2024.106766] [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: 06/20/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
Radiotherapy (RTx) is an essential and efficacious oncologic treatment, however, post-RTx bone fragility fractures present a challenging clinical problem. Cancer survivors treated with RTx are at variable risk for these late-onset, complex fragility fractures. Little data exists regarding the effects of RTx on bone fatigue properties despite the likelihood of fatigue loading as a mechanism leading up to atraumatic fracture. In this study, femurs collected from adult male rats were irradiated ex vivo with a therapeutic dose of x-irradiation (20 Gy), and then fatigued using a three-point bend setup. Femurs positioned in an isotonic bath at room temperature were loaded to a range of prescribed initial strain levels (based on beam theory equations, prior to any fatigue damage) at 3 Hz in force control. The goals of this study were to determine the feasibility of assessing RTx-induced alterations in 1) femur fatigue strength, 2) structural microdamage (creep and stiffness), and 3) tissue damage (diffuse damage and/or linear microcracking). Mid-diaphyseal morphology and tissue mineral density were not different between the RTx and Sham groups (p ≥ 0.35). With increasing applied apparent strain, the number of cycles to failure was reduced for the RTx femurs when compared to the Sham femurs (treatment x εapp, p = 0.041). RTx femurs had a greater phase II (steady state) creep rate (p = 0.0462) compared to Sham femurs. For femurs that reached 500k cycles, the RTx group had greater diffuse damage area (p = 0.015) than the Sham. This study provides evidence that radiation at therapeutic doses can directly diminish bone fatigue properties. This loss of fatigue properties is associated with increased structural fatigue damage and diffuse microdamage, without alterations in morphology or tissue mineral density, indicating a reduction in bone quality.
Collapse
Affiliation(s)
- Tara E Carney
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA; Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Amy E Biggs
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Mark A Miller
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Kenneth A Mann
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| |
Collapse
|
3
|
Pivonka P, Calvo-Gallego JL, Schmidt S, Martínez-Reina J. Advances in mechanobiological pharmacokinetic-pharmacodynamic models of osteoporosis treatment - Pathways to optimise and exploit existing therapies. Bone 2024; 186:117140. [PMID: 38838799 DOI: 10.1016/j.bone.2024.117140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/17/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
Osteoporosis (OP) is a chronic progressive bone disease which is characterised by reduction of bone matrix volume and changes in the bone matrix properties which can ultimately lead to bone fracture. The two major forms of OP are related to aging and/or menopause. With the worldwide increase of the elderly population, particularly age-related OP poses a serious health issue which puts large pressure on health care systems. A major challenge for development of new drug treatments for OP and comparison of drug efficacy with existing treatments is due to current regulatory requirements which demand testing of drugs based on bone mineral density (BMD) in phase 2 trials and fracture risk in phase 3 trials. This requires large clinical trials to be conducted and to be run for long time periods, which is very costly. This, together with the fact that there are already many drugs available for treatment of OP, makes the development of new drugs inhibitive. Furthermore, an increased trend of the use of different sequential drug therapies has been observed in OP management, such as sequential anabolic-anticatabolic drug treatment or switching from one anticatabolic drug to another. Running clinical trials for concurrent and sequential therapies is neither feasible nor practical due to large number of combinatorial possibilities. In silico mechanobiological pharmacokinetic-pharmacodynamic (PK-PD) models of OP treatments allow predictions beyond BMD, i.e. bone microdamage and degree of mineralisation can also be monitored. This will help to inform clinical drug usage and development by identifying the most promising scenarios to be tested clinically (confirmatory trials rather than exploratory only trials), optimise trial design and identify subgroups of the population that show benefit-risk profiles (both good and bad) that are different from the average patient. In this review, we provide examples of the predictive capabilities of mechanobiological PK-PD models. These include simulation results of PMO treatment with denosumab, implications of denosumab drug holidays and coupling of bone remodelling models with calcium and phosphate systems models that allows to investigate the effects of co-morbidities such as hyperparathyroidism and chronic kidney disease together with calcium and vitamin D status on drug efficacy.
Collapse
Affiliation(s)
- Peter Pivonka
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, QLD 4000, Australia.
| | - José Luis Calvo-Gallego
- Departmento de Ingeniería Mecánica y Fabricación, Universidad de Sevilla, Seville 41092, Spain
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA
| | - Javier Martínez-Reina
- Departmento de Ingeniería Mecánica y Fabricación, Universidad de Sevilla, Seville 41092, Spain
| |
Collapse
|
4
|
Elnunu IS, Redmond JN, Obata Y, Woolley W, Kammer DS, Acevedo C. Increased AGE Cross-Linking Reduces the Mechanical Properties of Osteons. JOM (WARRENDALE, PA. : 1989) 2024; 76:5692-5702. [PMID: 39318440 PMCID: PMC11417058 DOI: 10.1007/s11837-024-06716-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/10/2024] [Indexed: 09/26/2024]
Abstract
The osteon is the primary structural component of bone, contributing significantly to its unique toughness and strength. Despite extensive research on osteonal structure, the properties of osteons have not been fully investigated, particularly within the context of bone fragility diseases like type 2 diabetes mellitus (T2DM). This study aims to isolate osteons from bovine bone, simulate the effects of increased advanced glycation end-products (AGEs) in T2DM through ribosylation, and evaluate the mechanical properties of isolated osteons. Osteons extracted from the posterior section of bovine femur mid-diaphysis were processed to achieve a sub-millimeter scale for microscale imaging. Subsequently, synchrotron radiation micro-computed tomography was employed to precisely localize and isolate the osteon internally. While comparable elastic properties were observed between control and ribosylated osteons, the presence of AGEs led to decreased strain to failure. Young's modulus was quantified (9.9 ± 4.9 GPa and 8.7 ± 3 GPa, respectively), aligning closely with existing literature. This study presents a novel method for the extraction and isolation of osteons from bone and shows the detrimental effect of AGEs at the osteonal level. Supplementary Information The online version contains supplementary material available at 10.1007/s11837-024-06716-x.
Collapse
Affiliation(s)
- Ihsan S Elnunu
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT 84112 USA
| | - Jessica N Redmond
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT 84112 USA
| | - Yoshihiro Obata
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT 84112 USA
- Department of Mechanical and Aerospace Engineering, University of California San Diego, Engineers Ln, San Diego, CA 92161 USA
| | - William Woolley
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT 84112 USA
- Department of Mechanical and Aerospace Engineering, University of California San Diego, Engineers Ln, San Diego, CA 92161 USA
| | - David S Kammer
- Institute for Building Materials, ETH Zurich, Laura-Hezner-Weg 7, 8093 Zurich, Switzerland
| | - Claire Acevedo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT 84112 USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112 USA
- Department of Mechanical and Aerospace Engineering, University of California San Diego, Engineers Ln, San Diego, CA 92161 USA
| |
Collapse
|
5
|
Han Z, Gao X, Wang Y, Huang C, Song H, Cheng S, Yang X, Cui X, Wu J, Wei K, Cheng L. Autocatalytic bifunctional supramolecular hydrogels for osteoporotic bone repair. Natl Sci Rev 2024; 11:nwae209. [PMID: 39071098 PMCID: PMC11275467 DOI: 10.1093/nsr/nwae209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/01/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024] Open
Abstract
Conventional bone scaffolds, which are mainly ascribed to highly active osteoclasts and an inflammatory microenvironment with high levels of reactive oxygen species and pro-inflammatory factors, barely satisfy osteoporotic defect repair. Herein, multifunctional self-assembled supramolecular fiber hydrogels (Ce-Aln gel) consisting of alendronate (Aln) and cerium (Ce) ions were constructed for osteoporotic bone defect repair. Based on the reversible interaction and polyvalent cerium ions, the Ce-Aln gel, which was mainly composed of ionic coordination and hydrogen bonds, displayed good injectability and autocatalytic amplification of the antioxidant effect. In vitro studies showed that the Ce-Aln gel effectively maintained the biological function of osteoblasts by regulating redox homeostasis and improved the inflammatory microenvironment to enhance the inhibitory effect on osteoclasts. Ribonucleic acid (RNA) sequencing further revealed significant downregulation of various metabolic pathways, including apoptosis signaling, hypoxia metabolism and tumor necrosis factor-alpha (TNF-α) signaling via the nuclear factor kappa-B pathway after treatment with the Ce-Aln gel. In vivo experiments showed that the clinical drug-based Ce-Aln gel effectively promoted the tissue repair of osteoporotic bone defects by improving inflammation and inhibiting osteoclast formation at the defect. Notably, in vivo systemic osteoporosis was significantly ameliorated, highlighting the strong potential of clinical translation for precise therapy of bone defects.
Collapse
Affiliation(s)
- Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Xiang Gao
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Yuanjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Cheng Huang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Hao Song
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Shuning Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Xiaoyuan Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Xiaoliang Cui
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Jie Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Kailu Wei
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa 999078, China
| |
Collapse
|
6
|
Ravazzano L, Colaianni G, Tarakanova A, Xiao YB, Grano M, Libonati F. Multiscale and multidisciplinary analysis of aging processes in bone. NPJ AGING 2024; 10:28. [PMID: 38879533 PMCID: PMC11180112 DOI: 10.1038/s41514-024-00156-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/07/2024] [Indexed: 06/19/2024]
Abstract
The world population is increasingly aging, deeply affecting our society by challenging our healthcare systems and presenting an economic burden, thus turning the spotlight on aging-related diseases: exempli gratia, osteoporosis, a silent disease until you suddenly break a bone. The increase in bone fracture risk with age is generally associated with a loss of bone mass and an alteration in the skeletal architecture. However, such changes cannot fully explain increased fragility with age. To successfully tackle age-related bone diseases, it is paramount to comprehensively understand the fundamental mechanisms responsible for tissue degeneration. Aging mechanisms persist at multiple length scales within the complex hierarchical bone structure, raising the need for a multiscale and multidisciplinary approach to resolve them. This paper aims to provide an overarching analysis of aging processes in bone and to review the most prominent outcomes of bone aging. A systematic description of different length scales, highlighting the corresponding techniques adopted at each scale and motivating the need for combining diverse techniques, is provided to get a comprehensive description of the multi-physics phenomena involved.
Collapse
Affiliation(s)
- Linda Ravazzano
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Via Rubattino 81, Milano, 20134, Italy
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11, Bari, 70124, Italy
| | - Anna Tarakanova
- School of Mechanical, Aerospace, and Manufacturing Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, 06269, CT, USA
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, CT, 06269, Storrs, USA
| | - Yu-Bai Xiao
- School of Mechanical, Aerospace, and Manufacturing Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, 06269, CT, USA
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11, Bari, 70124, Italy
| | - Flavia Libonati
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Via Rubattino 81, Milano, 20134, Italy.
- Department of Mechanical, Energy, Management and Transport Engineering - DIME, University of Genova, Via all'Opera Pia 15, Genova, 16145, Italy.
| |
Collapse
|
7
|
Coppini A, Falconieri A, Mualem O, Nasrin SR, Roudon M, Saper G, Hess H, Kakugo A, Raffa V, Shefi O. Can repetitive mechanical motion cause structural damage to axons? Front Mol Neurosci 2024; 17:1371738. [PMID: 38912175 PMCID: PMC11191579 DOI: 10.3389/fnmol.2024.1371738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Biological structures have evolved to very efficiently generate, transmit, and withstand mechanical forces. These biological examples have inspired mechanical engineers for centuries and led to the development of critical insights and concepts. However, progress in mechanical engineering also raises new questions about biological structures. The past decades have seen the increasing study of failure of engineered structures due to repetitive loading, and its origin in processes such as materials fatigue. Repetitive loading is also experienced by some neurons, for example in the peripheral nervous system. This perspective, after briefly introducing the engineering concept of mechanical fatigue, aims to discuss the potential effects based on our knowledge of cellular responses to mechanical stresses. A particular focus of our discussion are the effects of mechanical stress on axons and their cytoskeletal structures. Furthermore, we highlight the difficulty of imaging these structures and the promise of new microscopy techniques. The identification of repair mechanisms and paradigms underlying long-term stability is an exciting and emerging topic in biology as well as a potential source of inspiration for engineers.
Collapse
Affiliation(s)
| | | | - Oz Mualem
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| | - Syeda Rubaiya Nasrin
- Graduate School of Science, Division of Physics and Astronomy, Kyoto University, Kyoto, Japan
| | - Marine Roudon
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Gadiel Saper
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Henry Hess
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Akira Kakugo
- Graduate School of Science, Division of Physics and Astronomy, Kyoto University, Kyoto, Japan
| | | | - Orit Shefi
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| |
Collapse
|
8
|
Ishizu H, Shimizu T, Arita K, Sato K, Takahashi R, Kusunoki K, Shimodan S, Asano T, Iwasaki N. Secondary fracture and mortality risk with very high fracture risk osteoporosis and proximal femoral fracture. J Bone Miner Metab 2024; 42:196-206. [PMID: 38308695 DOI: 10.1007/s00774-023-01492-1] [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: 09/24/2023] [Accepted: 12/06/2023] [Indexed: 02/05/2024]
Abstract
INTRODUCTION We aimed to investigate secondary fracture and mortality rates, and risk factors in patients with proximal femoral fractures. MATERIALS AND METHODS We conducted a multicenter prospective cohort study on female patients with proximal femoral fractures who underwent surgical treatment between April 2020 and March 2021. Postoperative follow-ups were performed at 6-, 12-, 18-, and 24-month intervals to determine the secondary fracture and mortality rates, and the risk factors and its influence were examined. RESULTS Of the 279 registered patients, 144 patients (51.6%) were diagnosed with very high fracture risk osteoporosis. The postoperative osteoporosis rate exceeded 96%; however, osteoanabolic agents were used sparingly. The risk factor of both secondary fracture and mortality was very high fracture risk osteoporosis, and secondary fractures within 12 months were markedly occurred. Secondary fracture rates increased as the number of matched very high fracture risk osteoporosis criteria increased. Notably, secondary fractures and mortality were recorded in 21.4% and 23.5% of the patients who met all criteria, respectively. CONCLUSION Over half of the female patients with proximal femoral fractures had very high fracture risk osteoporosis. Although, very high fracture risk osteoporosis demonstrated a notably increased risk of secondary fractures, particularly at 12 months post-surgery, the use of osteoanabolic agents was substantially low. Collectively, our findings highlight the need to consider the risk of very high fracture risk osteoporosis, expand the use of medications to include osteoanabolic agents, and reconsider the current healthcare approach for proximal femoral fractures.
Collapse
Affiliation(s)
- Hotaka Ishizu
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Tomohiro Shimizu
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Kosuke Arita
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
- Department of Orthopaedic Surgery, KKR Sapporo Medical Center, Sapporo, Japan
| | - Komei Sato
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
- Department of Orthopaedic Surgery, Iwamizawa City Hospital, Iwamizawa, Hokkaido, Japan
| | - Renya Takahashi
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
- Department of Orthopaedic Surgery, Kushiro City General Hospital, Kushiro, Hokkaido, Japan
| | - Kenichi Kusunoki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
- Central Hospital, Hakodate, Hokkaido, Japan
| | - Shun Shimodan
- Department of Orthopaedic Surgery, Kushiro City General Hospital, Kushiro, Hokkaido, Japan
| | - Tsuyoshi Asano
- Department of Orthopaedic Surgery, KKR Sapporo Medical Center, Sapporo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| |
Collapse
|
9
|
Jha SS, Srivastava A, Kambhampati SBS, Elhence A. Introduction to Osteoporosis, Osteomalacia, and Fragility Fractures. Indian J Orthop 2023; 57:25-32. [PMID: 38107821 PMCID: PMC10721584 DOI: 10.1007/s43465-023-01015-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 09/28/2023] [Indexed: 12/19/2023]
Abstract
Background Osteoporosis is a disease of the bones leading to decreased bone mineral density, leading to fragility fractures. This article is an overview of osteoporosis, osteomalacia and fragility fractures and serves as an introductory article for this special issue on osteoporosis. Methods This is a short, comprehensive account of the given conditions with concepts and a review from the recent literature. The authors provide relevant references from the literature in the bibliography. The sections herein have also been deliberated in the meetings of experts of osteoporosis. Results An overview of osteoporosis, osteomalacia and fragility fractures is provided, including definitions and summaries of aetiology, pathophysiology, diagnosis, prevention, and management. A detailed account of some of these topics will be provided in subsequent chapters. Conclusion Osteoporosis is a silent disease with the potential to cause significant morbidity and mortality if not detected early. It is important to differentiate from and diagnose associated osteomalacia to provide accurate therapy. It is also important to identify the type of fragility fractures and initiate treatment for bone strengthening to prevent subsequent fractures.
Collapse
|
10
|
Rohaiem SN, Khan BF, Al-Julaih GH, Mohammedin AS. Atraumatic Fractures in Multi-Morbid Older Adults: A Series of Five Cases and Review of Literature. Cureus 2023; 15:e51333. [PMID: 38288221 PMCID: PMC10824280 DOI: 10.7759/cureus.51333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2023] [Indexed: 01/31/2024] Open
Abstract
Atraumatic fractures (ATFs) are a fragility fracture subtype with occasional medicolegal issues. ATFs are defined as fractures because of a "low-energy mechanism that is usually considered incapable of producing a fracture." They are an underreported disorder, with epidemiological variations. ATF phenomena were previously reported not only in older adults, but also in children, young adults, older adults, and animals. This study is a short retrospective case series exploring atraumatic fractures in a tertiary care university hospital. Over a period of two years, a total of seven ATF cases were identified. However, only five fulfilled the inclusion criteria. Local causes of pathologic fractures (e.g., metastasis) and elder abuse or neglect were excluded. Comparison of the cases' clinical profile, fracture profile, and management was done. All five cases were frail females with significant osteotoxic burdens from medications and multi-morbidities. ATF presentations included typical (as pain) and atypical (as painless, loud crack, and sudden giveaway) symptomatology. One ATF had a coincident unexplained aseptic fever. Three cases had more than one fracture (fracture cascade), confirmed and followed up by x-rays. All the cases were managed conservatively except for one case that underwent hip hemiarthroplasty. Plans of care included managing the osteotoxic multi-morbidities burden, focusing on the whole body, not only on the fracture or bone. The study provided insights about challenges in presentations of ATF (as the bone fracture acute phase reaction: osteogenic aseptic fever). Risk factors are classically assumed to be osteoporosis, but it is usually systemic and multifactorial. A high risk of fracture warning sign could help decrease ATF occurrence or fracture cascades. Four ATF categories were detected to help healthcare systems identify high-risk patients and raise awareness among medical staff, families, and caregivers. Future studies of the at-risk groups are needed to understand ATF knowledge gaps, challenges, and the best treatments.
Collapse
Affiliation(s)
| | - Basim F Khan
- Pediatrics and Child Health, King Fahd Hospital of the University, Al-Khobar, SAU
| | | | - Ahmed S Mohammedin
- Geriatrics and Gerontology, Ain Shams University, Cairo, EGY
- Internal Medicine/Geriatrics, King Fahd Hospital of the University, Imam Abdulrahman bin Faisal University, Al-Khobar, SAU
| |
Collapse
|
11
|
Snow T, Woolley W, Acevedo C, Kingstedt OT. Effect of in vitro ribosylation on the dynamic fracture behavior of mature bovine cortical bone. J Mech Behav Biomed Mater 2023; 148:106171. [PMID: 37890344 DOI: 10.1016/j.jmbbm.2023.106171] [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: 11/01/2022] [Revised: 05/01/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023]
Abstract
In this study, the fracture behavior of ribosylated bovine cortical bone is investigated under loading conditions simulating a fall event. Single edge notched specimens, separated into a control group (n = 11) and a ribosylated group (n = 8), were extracted from the mid-diaphysis of a single bovine femur harvested from a mature cow. A seven-day ribosylation process results in the accumulation of Advanced-Glycation End Products (AGEs) cross-links and AGE adducts. Specimens were subjected to symmetric three point bending (opening mode) and an impact velocity of 1.6 m/s using a drop tower. Near-crack displacement fields up to fracture initiation are determined from high-speed images post-processed using digital image correlation. A constrained over-deterministic least squares regression and orthotropic material linear elastic fracture mechanics theory are used to extract the in-plane critical stress intensity factors at fracture initiation (i.e., fracture initiation toughness values). Statistically significant differences were not observed when comparing the in-plane fracture initiation toughness values (p≥0.96) or energy release rate (p=0.90) between the control and seven-day ribosylated groups. The intrinsic variability of bone may require high sample numbers in order to achieve an adequately powered experiment when assessing dynamic fracture behavior. While there are no detectable differences due to the ribosylation treatment investigated, this is likely due to the limited sample sizes utilized.
Collapse
Affiliation(s)
- Tanner Snow
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - William Woolley
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA; Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, 92093, USA
| | - Claire Acevedo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA; Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, 92093, USA.
| | - Owen T Kingstedt
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
12
|
Kamml J, Acevedo C, Kammer DS. Advanced-Glycation Endproducts: How cross-linking properties affect the collagen fibril behavior. J Mech Behav Biomed Mater 2023; 148:106198. [PMID: 37890341 PMCID: PMC11519298 DOI: 10.1016/j.jmbbm.2023.106198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
Advanced-Glycation-Endproducts (AGEs) are known to be a major cause of impaired tissue material properties. In collagen fibrils, which constitute a major building component of human tissue, these AGEs appear as fibrillar cross-links. It has been shown that when AGEs accumulate in collagen fibrils, a process often caused by diabetes and aging, the mechanical properties of the collagen fibril are altered. However, current knowledge about the mechanical properties of different types of AGEs, and their quantity in collagen fibrils is limited owing to the scarcity of available experimental data. Consequently, the precise relationship between the nano-scale cross-link properties, which differ from type to type, their density in collagen fibrils, and the mechanical properties of the collagen fibrils at larger scales remains poorly understood. In our study, we use coarse-grained molecular dynamics simulations and perform destructive tensile tests on collagen fibrils to evaluate the effect of different cross-link densities and their mechanical properties on collagen fibril deformation and fracture behavior. We observe that the collagen fibril stiffens at high strain levels when either the AGEs density or the loading energy capacity of AGEs are increased. Based on our results, we demonstrate that this stiffening is caused by a mechanism that favors energy absorption via stretching rather than inter-molecular sliding. Hence, in these cross-linked collagen fibrils, the absorbed energy is stored rather than dissipated through friction, resulting in brittle fracture upon fibrillar failure. Further, by varying multiple AGEs nano-scale parameters, we show that the AGEs loading energy capacity is, aside from their density in the fibril, the unique factor determining the effect of different types of AGEs on the mechanical behavior of collagen fibrils. Our results show that knowing AGEs properties is crucial for a better understanding of the nano-scale origin of impaired tissue behavior. We further suggest that future experimental investigations should focus on the quantification of the loading energy capacity of AGEs as a key property for their influence on collagen fibrils.
Collapse
Affiliation(s)
- Julia Kamml
- Institute for Building Materials, ETH Zurich, Switzerland
| | - Claire Acevedo
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
| | - David S Kammer
- Institute for Building Materials, ETH Zurich, Switzerland.
| |
Collapse
|
13
|
Lee SS, Kleger N, Kuhn GA, Greutert H, Du X, Smit T, Studart AR, Ferguson SJ. A 3D-Printed Assemblable Bespoke Scaffold as a Versatile Microcryogel Carrier for Site-Specific Regenerative Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302008. [PMID: 37632210 DOI: 10.1002/adma.202302008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/22/2023] [Indexed: 08/27/2023]
Abstract
Advances in additive manufacturing have led to diverse patient-specific implant designs utilizing computed tomography, but this requires intensive work and financial implications. Here, Digital Light Processing is used to fabricate a hive-structured assemblable bespoke scaffold (HIVE). HIVE can be manually assembled in any shape/size with ease, so a surgeon can create a scaffold that will best fit a defect before implantation. Simultaneously, it can have site-specific treatments by working as a carrier filled with microcryogels (MC) incorporating different biological factors in different pockets of HIVE. After characterization, possible site-specific applications are investigated by utilizing HIVE as a versatile carrier with incorporated treatments such as growth factors (GF), bioceramic, or cells. HIVE as a GF-carrier shows a controlled release of bone morphogenetic protein/vascular endothelial growth factor (BMP/VEGF) and induced osteogenesis/angiogenesis from human mesenchymal stem cells (hMSC)/human umbilical vein endothelial cells (HUVECs). Furthermore, as a bioceramic-carrier, HIVE demonstrates enhanced mineralization and osteogenesis, and as a HUVEC carrier, it upregulates both osteogenic and angiogenic gene expression of hMSCs. HIVE with different combinations of MCs yields a distinct local effect and successful cell migration is confirmed within assembled HIVEs. Finally, an in vivo rat subcutaneous implantation demonstrates site-specific osteogenesis and angiogenesis.
Collapse
Affiliation(s)
- Seunghun S Lee
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Nicole Kleger
- Complex Materials, Department of Materials, ETH Zurich, Zurich, 8093, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Helen Greutert
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Xiaoyu Du
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Thijs Smit
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - André R Studart
- Complex Materials, Department of Materials, ETH Zurich, Zurich, 8093, Switzerland
| | - Stephen J Ferguson
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| |
Collapse
|
14
|
Maghami E, Najafi A. Microstructural fatigue fracture behavior of glycated cortical bone. Med Biol Eng Comput 2023; 61:3021-3034. [PMID: 37582979 DOI: 10.1007/s11517-023-02901-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 07/29/2023] [Indexed: 08/17/2023]
Abstract
The current study aims to simulate fatigue microdamage accumulation in glycated cortical bone with increased advanced glycation end-products (AGEs) using a phase field fatigue framework. We link the material degradation in the fracture toughness of cortical bone to the high levels of AGEs in this tissue. We simulate fatigue fracture in 2D models of cortical bone microstructure extracted from human tibias. The results present that the mismatch between the critical energy release rate of microstructural features (e.g., osteons and interstitial tissue) can alter crack initiation and propagation patterns. Moreover, the high AGEs content through the increased mismatch ratio can cause the activation or deactivation of bone toughening mechanisms under cyclic loading. The fatigue fracture simulations also show that the lifetime of diabetic cortical bone samples can be dependent on the geometry of microstructural features and the mismatch ratio between the features. Additionally, the results indicate that the trapped cracks in cement lines in the diabetic cortical microstructure can prevent further crack growth under cyclic loading. The present findings show that alterations in the materials heterogeneity of microstructural features can change the fatigue fracture response, lifetime, and fragility of cortical bone with high AGEs contents. Cortical bone models are created from microscopy images taken from the cortical cross-section of human tibias. Increased glycation contents in the cortical bone sample can change the crack growth trajectories.
Collapse
|
15
|
Wang B, Vashishth D. Advanced glycation and glycoxidation end products in bone. Bone 2023; 176:116880. [PMID: 37579812 PMCID: PMC10529863 DOI: 10.1016/j.bone.2023.116880] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/21/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Hyperglycemia and oxidative stress, enhanced in diabetes and aging, result in excessive accumulation of advanced glycation and glycoxidation end products (AGEs/AGOEs) in bone. AGEs/AGOES are considered to be "the missing link" in explaining increased skeletal fragility with diabetes, aging, and osteoporosis where increased fracture risk cannot be solely explained by bone mass and/or fall incidences. AGEs/AGOEs disrupt bone turnover and deteriorate bone quality through alterations of organic matrix (collagen and non-collagenous proteins), mineral, and water content. AGEs and AGOEs are also associated with bone fragility in other conditions such as Alzheimer's disease, circadian rhythm disruption, and cancer. This review explains how AGEs and AGOEs accumulate in bone and impact bone quality and bone fracture, and how AGES/AGOEs are being targeted in preclinical and clinical investigations for inhibition or removal, and for prediction and management of diabetic, osteoporotic and insufficiency fractures.
Collapse
Affiliation(s)
- Bowen Wang
- Shirley Ann Jackson Ph.D. Center of Biotechnology and Interdisciplinary Studies, Troy, NY 12180, USA; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Deepak Vashishth
- Shirley Ann Jackson Ph.D. Center of Biotechnology and Interdisciplinary Studies, Troy, NY 12180, USA; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA; Rensselaer - Icahn School of Medicine at Mount Sinai Center for Engineering and Precision Medicine, New York, NY 10019, USA.
| |
Collapse
|
16
|
Hu M, Zhang Y, Guo J, Guo C, Yang X, Ma X, Xu H, Xiang S. Meta-analysis of the effects of denosumab and romosozumab on bone mineral density and turnover markers in patients with osteoporosis. Front Endocrinol (Lausanne) 2023; 14:1188969. [PMID: 37529613 PMCID: PMC10390296 DOI: 10.3389/fendo.2023.1188969] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/14/2023] [Indexed: 08/03/2023] Open
Abstract
Purpose To assess the alterations in bone mineral density and bone turnover marker concentrations following the administration of denosumab and romosozumab therapies in patients with osteoporosis. Methods PubMed was searched for studies published until January 28, 2023, that investigated the clinical efficacy and bone turnover marker changes of denosumab and romosozumab in the treatment of osteoporosis, with a minimum follow-up of 3 months in each study. Studies were screened, and data on changes in bone mineral density (BMD), P1NP, and TRACP-5b levels after treatment were extracted and included in the analysis. Results Six studies were analyzed. At 3 months after treatment, the romosozumab group showed greater changes in lumbar BMD and bone turnover markers. BMD of total hip and femoral neck was relatively delayed. Beginning at 6 to 12 months, romosozumab showed greater changes in bone mineral density and markers of bone turnover. Conclusion Both romosozumab and denosumab have antiosteoporotic effects, with greater effects on BMD and bone turnover markers observed within 12 months of romosozumab treatment. Systematic Review Registration https://www.crd.york.ac.uk/prospero, identifier CRD42023395034.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hao Xu
- *Correspondence: Hao Xu, ; Shuai Xiang,
| | | |
Collapse
|
17
|
Kamml J, Ke CY, Acevedo C, Kammer DS. The influence of AGEs and enzymatic cross-links on the mechanical properties of collagen fibrils. J Mech Behav Biomed Mater 2023; 143:105870. [PMID: 37156073 PMCID: PMC11522032 DOI: 10.1016/j.jmbbm.2023.105870] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/28/2023] [Accepted: 04/23/2023] [Indexed: 05/10/2023]
Abstract
Collagen, one of the main building blocks for various tissues, derives its mechanical properties directly from its structure of cross-linked tropocollagen molecules. The cross-links are considered to be a key component of collagen fibrils as they can change the fibrillar behavior in various ways. For instance, enzymatic cross-links (ECLs), one particular type of cross-links, are known for stabilizing the structure of the fibril and improving material properties, while cross-linking AGEs (Advanced-Glycation Endproducts) have been shown to accumulate and impair the mechanical properties of collageneous tissues. However, the reasons for whether and how a given type of cross-link improves or impairs the material properties remain unknown, and the exact relationship between the cross-link properties and density, and the fibrillar behavior is still not well understood. Here, we use coarse-grained steered molecular models to evaluate the effect of AGEs and ECLs cross-links content on the deformation and failure properties of collagen fibrils. Our simulations show that the collagen fibrils stiffen at high strain levels when the AGEs content exceeds a critical value. In addition, the strength of the fibril increases with AGEs accumulation. By analyzing the forces within the different types of cross-links (AGEs and ECLs) as well as their failure, we demonstrate that a change of deformation mechanism is at the origin of these observations. A high AGEs content reinforces force transfer through AGEs cross-links rather than through friction between sliding tropocollagen molecules, which leads to failure by breaking of bonds within the tropocollagen molecules. We show that this failure mechanism, which is associated with lower energy dissipation, results in more abrupt failure of the collagen fibril. Our results provide a direct and causal link between increased AGEs content, inhibited intra-fibrillar sliding, increased stiffness, and abrupt fibril fracture. Therefore, they explain the mechanical origin of bone brittleness as commonly observed in elderly and diabetic populations. Our findings contribute to a better understanding of the mechanisms underlying impaired tissue behavior due to elevated AGEs content and could enable targeted measures regarding the reduction of specific collagen cross-linking levels.
Collapse
Affiliation(s)
- Julia Kamml
- Institute for Building Materials, ETH Zurich, Switzerland
| | - Chun-Yu Ke
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA, USA
| | - Claire Acevedo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - David S Kammer
- Institute for Building Materials, ETH Zurich, Switzerland.
| |
Collapse
|
18
|
Li W, Wu Y, Zhang X, Wu T, Huang K, Wang B, Liao J. Self-healing hydrogels for bone defect repair. RSC Adv 2023; 13:16773-16788. [PMID: 37283866 PMCID: PMC10240173 DOI: 10.1039/d3ra01700a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/24/2023] [Indexed: 06/08/2023] Open
Abstract
Severe bone defects can be caused by various factors, such as tumor resection, severe trauma, and infection. However, bone regeneration capacity is limited up to a critical-size defect, and further intervention is required. Currently, the most common clinical method to repair bone defects is bone grafting, where autografts are the "gold standard." However, the disadvantages of autografts, including inflammation, secondary trauma and chronic disease, limit their application. Bone tissue engineering (BTE) is an attractive strategy for repairing bone defects and has been widely researched. In particular, hydrogels with a three-dimensional network can be used as scaffolds for BTE owing to their hydrophilicity, biocompatibility, and large porosity. Self-healing hydrogels respond rapidly, autonomously, and repeatedly to induced damage and can maintain their original properties (i.e., mechanical properties, fluidity, and biocompatibility) following self-healing. This review focuses on self-healing hydrogels and their applications in bone defect repair. Moreover, we discussed the recent progress in this research field. Despite the significant existing research achievements, there are still challenges that need to be addressed to promote clinical research of self-healing hydrogels in bone defect repair and increase the market penetration.
Collapse
Affiliation(s)
- Weiwei Li
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Yanting Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Xu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Tingkui Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Kangkang Huang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Beiyu Wang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| |
Collapse
|
19
|
Wang T, Liu K, Wang J, Xiang G, Hu X, Bai H, Lei W, Tao TH, Feng Y. Spatiotemporal Regulation of Injectable Heterogeneous Silk Gel Scaffolds for Accelerating Guided Vertebral Repair. Adv Healthc Mater 2023; 12:e2202210. [PMID: 36465008 DOI: 10.1002/adhm.202202210] [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: 08/30/2022] [Revised: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Osteoporotic vertebral fracture is jeopardizing the health of the aged population around the world, while the hypoxia microenvironment and oxidative damage of bone defect make it difficult to perform effective tissue regeneration. The balance of oxidative stress and the coupling of vessel and bone ingrowth are critical for bone regeneration. In this study, an injectable heterogeneous silk gel scaffold which can spatiotemporally and sustainedly release bone mesenchymal stem cell-derived small extracellular vesicles, HIF-1α pathway activator, and inhibitor is developed for bone repair and vertebral reinforcement. The initial enhancement of HIF-1α upregulates the expression of VEGF to promote angiogenesis, and the balance of reactive oxygen species level is regulated to effectively eliminate oxidative damage and abnormal microenvironment. The subsequent inhibition of HIF-1α avoids the overexpression of VEGF and vascular overgrowth. Meanwhile, complex macroporous structures and suitable mechanical support can be obtained within the silk gel scaffolds, which will promote in situ bone regeneration. These findings provide a new clinical translation strategy for osteoporotic vertebral augmentation on basis of hypoxia microenvironment improvement.
Collapse
Affiliation(s)
- Tianji Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Keyin Liu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Jing Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Geng Xiang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaofan Hu
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Bai
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Tiger H Tao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.,School of Physical Science and Technology, ShanghaiTech University, Shanghai, 200031, China.,Institute of Brain-Intelligence Technology, Zhangjiang Laboratory, Shanghai, 200031, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 200031, China
| | - Yafei Feng
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
20
|
Lee J, Kim D, Park S, Baek S, Jung J, Kim T, Han DK. Nitric Oxide-Releasing Bioinspired Scaffold for Exquisite Regeneration of Osteoporotic Bone via Regulation of Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205336. [PMID: 36581472 PMCID: PMC9951336 DOI: 10.1002/advs.202205336] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Osteoporotic bone regeneration is a challenging process which involves the occurrence of sophisticated interactions. Although various polymeric scaffolds have been proposed for bone repair, research on osteoporotic bone regeneration remains practically limited. In particular, achieving satisfactory bone regeneration when using osteoporotic drugs is challenging including bisphosphonates. Here, a novel nitric oxide-releasing bioinspired scaffold with bioactive agents for the exquisite regeneration of osteoporotic bone is proposed. The bone-like biomimetic poly(lactic-co-glycolic acid) scaffold is first prepared in combination with organic/inorganic ECM and magnesium hydroxide as the base implant material. Nanoparticles containing bioactive agents of zinc oxide (ZO), alendronate, and BMP2 are incorporated to the biomimetic scaffold to impart multifunctionality such as anti-inflammation, angiogenesis, anti-osteoclastogenesis, and bone regeneration. Especially, nitric oxide (NO) generated from ZO stimulates the activity of cGMP and protein kinase G; in addition, ZO downregulates the RANKL/osteoprotegerin ratio by suppressing the Wnt/β-catenin signaling pathway. The new bone is formed much better in the osteoporotic rat model than in the normal model through the regulation of bone homeostasis via the scaffold. These synergistic effects suggest that such a bioinspired scaffold could be a comprehensive way to regenerate exceptionally osteoporotic bones.
Collapse
Affiliation(s)
- Jun‐Kyu Lee
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| | - Da‐Seul Kim
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- School of Integrative EngineeringChung‐Ang University84 Heukseok‐ro, Dongjak‐guSeoul06974Republic of Korea
| | - So‐Yeon Park
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- Division of BiotechnologyCollege of Life Sciences and BiotechnologyKorea UniversitySeongbuk‐guSeoul02841Republic of Korea
| | - Seung‐Woon Baek
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- Department of Biomedical EngineeringSKKU Institute for ConvergenceSungkyunkwan University (SKKU)2066 Seobu‐ro, Jangan‐gu, Suwon‐siGyeonggi‐do16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSKKU Institute for ConvergenceSungkyunkwan University (SKKU)2066 Seobu‐ro, Jangan‐gu, Suwon‐siGyeonggi‐do16419Republic of Korea
| | - Ji‐Won Jung
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| | - Tae‐Hyung Kim
- School of Integrative EngineeringChung‐Ang University84 Heukseok‐ro, Dongjak‐guSeoul06974Republic of Korea
| | - Dong Keun Han
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| |
Collapse
|
21
|
Sacher SE, Hunt HB, Lekkala S, Lopez KA, Potts J, Heilbronner AK, Stein EM, Hernandez CJ, Donnelly E. Distributions of Microdamage Are Altered Between Trabecular Rods and Plates in Cancellous Bone From Men With Type 2 Diabetes Mellitus. J Bone Miner Res 2022; 37:740-752. [PMID: 35064941 PMCID: PMC9833494 DOI: 10.1002/jbmr.4509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 01/13/2023]
Abstract
Individuals with type 2 diabetes mellitus (T2DM) have an increased risk of fragility fracture despite exhibiting normal to high bone mineral density (BMD). Conditions arising from T2DM, such as reduced bone turnover and alterations in microarchitecture, may contribute to skeletal fragility by influencing bone morphology and microdamage accumulation. The objectives of this study were (i) to characterize the effect of T2DM on microdamage quantity and morphology in cancellous bone, and (ii) relate the accumulation of microdamage to the cancellous microarchitecture. Cancellous specimens from the femoral neck were collected during total hip arthroplasty (T2DM: n = 22, age = 65 ± 9 years, glycated hemoglobin [HbA1c] = 7.00% ± 0.98%; non-diabetic [non-DM]: n = 25, age = 61 ± 8 years, HbA1c = 5.50% ± 0.4%), compressed to 3% strain, stained with lead uranyl acetate to isolate microdamage, and scanned with micro-computed tomography (μCT). Individual trabeculae segmentation was used to isolate rod-like and plate-like trabeculae and their orientations with respect to the loading axis. The T2DM group trended toward a greater BV/TV (+27%, p = 0.07) and had a more plate-like trabecular architecture (+8% BVplates , p = 0.046) versus non-DM specimens. Rods were more damaged relative to their volume compared to plates in the non-DM group (DVrods /BVrods versus DVplates /BVplates : +49%, p < 0.0001), but this difference was absent in T2DM specimens. Longitudinal rods were more damaged in the non-DM group (DVlongitudinal rods /BVlongitudinal rods : +73% non-DM versus T2DM, p = 0.027). Total damage accumulation (DV/BV) and morphology (DS/DV) did not differ in T2DM versus non-DM specimens. These results provide evidence that cancellous microarchitecture does not explain fracture risk in T2DM, pointing to alterations in material matrix properties. In particular, cancellous bone from men with T2DM may have an attenuated ability to mitigate microdamage accumulation through sacrificial rods. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Sara E Sacher
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
| | - Heather B Hunt
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
| | - Sashank Lekkala
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
| | - Kelsie A Lopez
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
| | - Jesse Potts
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
| | - Alison K Heilbronner
- Department of Medicine, Endocrinology and Metabolic Bone Service, Hospital for Special Surgery, New York, NY, USA
| | - Emily M Stein
- Department of Medicine, Endocrinology and Metabolic Bone Service, Hospital for Special Surgery, New York, NY, USA
| | - Christopher J Hernandez
- Research Division, Hospital for Special Surgery, New York, NY, USA.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Eve Donnelly
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA.,Research Division, Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
22
|
Domínguez-Zotes S, Fuertes MA, Rodríguez-Huete A, Valbuena A, Mateu MG. A Genetically Engineered, Chain Mail-Like Nanostructured Protein Material with Increased Fatigue Resistance and Enhanced Self-Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105456. [PMID: 35060301 DOI: 10.1002/smll.202105456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/10/2021] [Indexed: 06/14/2023]
Abstract
Protein-based nanostructured materials are being developed for many biomedical and nanotechnological applications. Despite their many desirable features, protein materials are highly susceptible to disruption by mechanical stress and fatigue. This study is aimed to increase fatigue resistance and enhance self-healing of a natural protein-based supramolecular nanomaterial through permanent genetic modification. The authors envisage the conversion of a model nanosheet, formed by a regular array of noncovalently bound human immunodeficiency virus capsid protein molecules, into a supramolecular "chain mail." Rationally engineered mutations allow the formation of a regular network of disulfide bridges in the protein lattice. This network links each molecule in the lattice to each adjacent molecule through one covalent bond, analogous to the rivetting of interlinked iron rings in the chain mail of a medieval knight. The engineered protein nanosheet shows greatly increased thermostability and resistance to mechanical stress and fatigue in particular, as well as enhanced self-healing, without undesirable stiffening compared to the original material. The results provide proof of concept for a genetic design to permanently increase fatigue resistance and enhance self-healing of protein-based nanostructured materials. They also provide insights into the molecular basis for fatigue of protein materials.
Collapse
Affiliation(s)
- Santos Domínguez-Zotes
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Cantoblanco, Madrid, 28049, Spain
| | - Miguel Angel Fuertes
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Cantoblanco, Madrid, 28049, Spain
| | - Alicia Rodríguez-Huete
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Cantoblanco, Madrid, 28049, Spain
| | - Alejandro Valbuena
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Cantoblanco, Madrid, 28049, Spain
| | - Mauricio G Mateu
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Cantoblanco, Madrid, 28049, Spain
| |
Collapse
|
23
|
Ma Y, Ran D, Zhao H, Shi X, Song R, Zou H, Liu Z. The effect of P2X7R- mediated Ca 2+ and MAPK signaling in OPG-induced duck embryo osteoclasts differentiation and adhesive structure damage. Life Sci 2022; 293:120337. [PMID: 35074408 DOI: 10.1016/j.lfs.2022.120337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/10/2022] [Accepted: 01/15/2022] [Indexed: 10/19/2022]
Abstract
Various factors cause animal bone malnutrition disease during intensive culture. Osteoclasts play an important role in regulating bone metabolism disease. Osteoprotegerin (OPG) modulates osteoclast function; however, the mechanism underlying this effect is unknown. Therefore, the present study aimed to explore whether OPG affects duck embryo osteoclast function via purinergic receptor P2X7. OPG significantly inhibited duck embryo osteoclast differentiation and bone resorption, and suppressed F-actin formation. In addition, OPG remarkably impaired duck embryo osteoclasts' adhesive structure. After OPG treatment, the expression of P2X7R significantly reduced, the ATP level and Ca2+-ATPase activity decreased rapidly, and concomitantly suppressed calcium and MAPK signaling. A438079 (a selective P2X7R inhibitor) significantly inhibited duck embryo osteoclast differentiation and bone resorption, and the phosphorylation of Ca2+ regulated proteins (CAM, CAMKII, CAMKIV) and MAPKs (ERK, JNK, and P38) were markedly suppressed. Pretreatment of duck embryo osteoclasts with BzATP, a P2X7R agonist, activated Ca2+ and MAPK signaling. BzATP alleviated OPG-induced duck embryo osteoclast differentiation and adhesive structure damage, and recovered the distribution of adhesion-related proteins in mature duck embryo osteoclasts. Thus, P2RX7-mediated Ca2+ and MAPK signaling has a key function in OPG-induced duck embryo osteoclast differentiation and adhesive structure damage. P2X7R might be an ideal target to treat bone diseases through regulating bone cell activation.
Collapse
Affiliation(s)
- Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Di Ran
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Hongyan Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Xueni Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| |
Collapse
|
24
|
Lee SS, Laganenka L, Du X, Hardt WD, Ferguson SJ. Silicon Nitride, a Bioceramic for Bone Tissue Engineering: A Reinforced Cryogel System With Antibiofilm and Osteogenic Effects. Front Bioeng Biotechnol 2021; 9:794586. [PMID: 34976982 PMCID: PMC8714913 DOI: 10.3389/fbioe.2021.794586] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 01/05/2023] Open
Abstract
Silicon nitride (SiN [Si3N4]) is a promising bioceramic for use in a wide variety of orthopedic applications. Over the past decades, it has been mainly used in industrial applications, such as space shuttle engines, but not in the medical field due to scarce data on the biological effects of SiN. More recently, it has been increasingly identified as an emerging material for dental and orthopedic implant applications. Although a few reports about the antibacterial properties and osteoconductivity of SiN have been published to date, there have been limited studies of SiN-based scaffolds for bone tissue engineering. Here, we developed a silicon nitride reinforced gelatin/chitosan cryogel system (SiN-GC) by loading silicon nitride microparticles into a gelatin/chitosan cryogel (GC), with the aim of producing a biomimetic scaffold with antibiofilm and osteogenic properties. In this scaffold system, the GC component provides a hydrophilic and macroporous environment for cells, while the SiN component not only provides antibacterial properties and osteoconductivity but also increases the mechanical stiffness of the scaffold. This provides enhanced mechanical support for the defect area and a better osteogenic environment. First, we analyzed the scaffold characteristics of SiN-GC with different SiN concentrations, followed by evaluation of its apatite-forming capacity in simulated body fluid and protein adsorption capacity. We further confirmed an antibiofilm effect of SiN-GC against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) as well as enhanced cell proliferation, mineralization, and osteogenic gene upregulation for MC3T3-E1 pre-osteoblast cells. Finally, we developed a bioreactor to culture cell-laden scaffolds under cyclic compressive loading to mimic physiological conditions and were able to demonstrate improved mineralization and osteogenesis from SiN-GC. Overall, we confirmed the antibiofilm and osteogenic effect of a silicon nitride reinforced cryogel system, and the results indicate that silicon nitride as a biomaterial system component has a promising potential to be developed further for bone tissue engineering applications.
Collapse
Affiliation(s)
- Seunghun S. Lee
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Leanid Laganenka
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Xiaoyu Du
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Stephen J. Ferguson
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
25
|
Wang Z, Liu Y, Zhang J, Lin M, Xiao C, Bai H, Liu C. Mechanical loading alleviated the inhibition of β2-adrenergic receptor agonist terbutaline on bone regeneration. FASEB J 2021; 35:e22033. [PMID: 34739146 DOI: 10.1096/fj.202101045rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022]
Abstract
The long-term use of adrenergic medication in treating various conditions, such as asthma, increases the chances of bone fracture. Dynamic mechanical loading at a specific time is a method for improving bone quality and promoting healing. Therefore, we hypothesized that precisely controlling the mechanical environment can contribute to the alleviation of the negative effects of chronic treatment with the common asthma drug terbutaline, which is a β2-adrenergic receptor agonist that facilitates bone homeostasis and defect repair through its anabolic effect on osteogenic cells. Our in vitro results showed that terbutaline can directly inhibit osteogenesis by impairing osteogenic differentiation and mineralization. Chronic treatment in vivo was simulated by administering terbutaline to C57BL/6J mice for 4 weeks before bone defect surgery and mechanical loading. We utilized a stabilized tibial defect model, which allowed the application of anabolic mechanical loading. During homeostasis, chronic terbutaline treatment reduced the bone formation rate, the fracture toughness of long bones, and the concentrations of bone formation markers in the sera. During defect repair, terbutaline decreased the bone volume, type H vessel, and total blood vessel volume. Terbutaline treatment reduced the number of osteogenic cells. Periostin, which was secreted mainly by Prrx1+ osteoprogenitors and F4/80+ macrophages, was inhibited by treating the bone defect with terbutaline. Interestingly, controlled mechanical loading facilitated the recovery of bone volume and periostin expression and the number of osteogenic cells within the defect. In conclusion, mechanical loading can rescue negative effects on new bone accrual and repair induced by chronic terbutaline treatment.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jianing Zhang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chufan Xiao
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Haoying Bai
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
26
|
Estrogen depletion alters osteogenic differentiation and matrix production by osteoblasts in vitro. Exp Cell Res 2021; 408:112814. [PMID: 34492267 DOI: 10.1016/j.yexcr.2021.112814] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/20/2021] [Accepted: 09/03/2021] [Indexed: 11/21/2022]
Abstract
Recent studies have revealed that the effects of estrogen deficiency are not restricted to osteoclasts and bone resorption, but that bone matrix composition is altered and osteoblasts exhibit an impaired response to mechanical stimulation. In this study, we test the hypothesis that estrogen depletion alters osteogenic differentiation and matrix production by mechanically stimulated osteoblasts in vitro. MC3T3-E1 cells were pre-treated with estrogen for 14 days, after which estrogen was withdrawn or inhibited with Fulvestrant up to 14 days. Fluid shear stress (FSS) was applied using an orbital shaker. Under estrogen depletion in static culture, osteogenic marker (ALP) and gene expression (Runx2) were decreased at 2 and after 7 days of estrogen depletion, respectively. In addition, up to 7 day the inhibition of the estrogen receptor significantly decreased fibronectin expression (FN1) under static conditions. Under estrogen depletion and daily mechanical stimulation, changes in expression of Runx2 occurred earlier (4 days) and by 14 days, changes in matrix production (Col1a1) were reported. We propose that changes in osteoblast differentiation and impaired matrix production during estrogen depletion may contribute to the altered quality of the bone and act as a contributing factor to increased bone fragility in postmenopausal osteoporosis.
Collapse
|
27
|
Migliorini F, Giorgino R, Hildebrand F, Spiezia F, Peretti GM, Alessandri-Bonetti M, Eschweiler J, Maffulli N. Fragility Fractures: Risk Factors and Management in the Elderly. MEDICINA-LITHUANIA 2021; 57:medicina57101119. [PMID: 34684156 PMCID: PMC8538459 DOI: 10.3390/medicina57101119] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 02/08/2023]
Abstract
Given the progressive ageing of Western populations, the fragility fractures market has a growing socioeconomic impact. Fragility fractures are common in the elderly, negatively impacting their quality of life, limiting autonomy, increasing disability, and decreasing life expectancy. Different causes contribute to the development of a fractures in frail individuals. Among all, targeting fragile patients before the development of a fracture may represent the greatest challenge, and current diagnostic tools suffer from limitations. This study summarizes the current evidence on the management of fragility fractures, discussing risk factors, prevention, diagnosis, and actual limitations of the clinical therapeutic options, putting forward new ideas for further scientific investigation.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedics, Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany; (F.M.); (F.H.); (J.E.)
| | - Riccardo Giorgino
- Residency Program in Orthopedics and Traumatology, University of Milan, 20122 Milan, Italy;
| | - Frank Hildebrand
- Department of Orthopaedics, Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany; (F.M.); (F.H.); (J.E.)
| | - Filippo Spiezia
- Department of Orthopaedic and Trauma Surgery, Ospedale San Carlo Potenza, Via Potito Petrone, 85100 Potenza, Italy;
| | - Giuseppe Maria Peretti
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
- IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy
| | | | - Jörg Eschweiler
- Department of Orthopaedics, Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany; (F.M.); (F.H.); (J.E.)
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Thornburrow Drive, Stoke-on-Trent ST4 7QB, UK
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Mile End Hospital, London E1 4DG, UK
- Correspondence:
| |
Collapse
|
28
|
Li Z, Xiang S, Lin Z, Li EN, Yagi H, Cao G, Yocum L, Li L, Hao T, Bruce KK, Fritch MR, Hu H, Wang B, Alexander PG, Khor KA, Tuan RS, Lin H. Graphene oxide-functionalized nanocomposites promote osteogenesis of human mesenchymal stem cells via enhancement of BMP-SMAD1/5 signaling pathway. Biomaterials 2021; 277:121082. [PMID: 34464823 DOI: 10.1016/j.biomaterials.2021.121082] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/07/2021] [Accepted: 08/21/2021] [Indexed: 12/28/2022]
Abstract
Biomaterials that can harness the intrinsic osteogenic potential of stem cells offer a promising strategy to accelerate bone regeneration and repair. Previously, we had used methacrylated gelatin (GelMA)-based scaffolds to achieve bone formation from human mesenchymal stem cells (hMSCs). In this study, we aimed to further enhance hMSC osteogenesis by incorporating graphene oxide (GO)-based nanosheets into GelMA. In vitro results showed high viability and metabolic activities in hMSCs encapsulated in the newly developed nanocomposites. Incorporation of GO markedly increased mineralization within hMSC-laden constructs, which was further increased by replacing GO with silica-coated graphene oxide (SiGO). Mechanistic analysis revealed that the nanosheet enhanced the production, retention, and biological activity of endogenous bone morphogenetic proteins (BMPs), resulting in robust osteogenesis in the absence of exogenous osteoinductive growth factors. Specifically, the osteoinductive effect of the nanosheets was abolished by inhibiting the BMP signaling pathway with LDN-193189 treatment. The bone formation potential of the technology was further tested in vivo using a mouse subcutaneous implantation model, where hMSCs-laden GO/GelMA and SiGO/GelMA samples resulted in bone volumes 108 and 385 times larger, respectively, than the GelMA control group. Taken together, these results demonstrate the biological activity and mechanism of action of GO-based nanosheets in augmenting the osteogenic capability of hMSCs, and highlights the potential of leveraging nanomaterials such as GO and SiGO for bone tissue engineering applications.
Collapse
Affiliation(s)
- Zhong Li
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shiqi Xiang
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zixuan Lin
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eileen N Li
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Haruyo Yagi
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guorui Cao
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Yocum
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - La Li
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tingjun Hao
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katherine K Bruce
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Madalyn R Fritch
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Huanlong Hu
- School of Mechanical & Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Bing Wang
- Molecular Therapeutics Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter G Alexander
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Khiam Aik Khor
- School of Mechanical & Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Rocky S Tuan
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Hang Lin
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Sacher S, Hernandez CJ, Donnelly E. Characterization of Ultralow Density Cellular Solids: Lessons from 30 years of Bone Biomechanics Research. ADVANCED ENGINEERING MATERIALS 2021; 23:2100206. [PMID: 34456625 PMCID: PMC8389487 DOI: 10.1002/adem.202100206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Indexed: 06/13/2023]
Abstract
Advances in additive manufacturing techniques have enabled the development of micro-architectured materials displaying a combination of low-density and lightweight structures with high specific strength and toughness. The mechanical performance of micro-architectured materials can be assessed using standard techniques; however, when studying low- and ultralow density micro-architectured materials, standard characterization techniques can be subject to experimental artifacts. Additionally, quantitative assessment and comparisons of microarchitectures with distinct lattice patterns is not always straightforward. Cancellous bone is a natural, ultralow density (porosity often exceeding 90%), irregular, cellular solid that has been thoroughly characterized in terms of micro-architecture and mechanical performance over the past 30 years. However, most the literature on cancellous bone mechanical properties and micro-structure-function relationships is in the medical literature and is not immediately accessible to materials designers. Here we provide a brief review of state-of-the-art approaches for characterizing the micro-architecture and mechanical performance of ultralow density cancellous bone, including methods of addressing experimental artifacts during mechanical characterization of ultralow density cellular solids, methods of quantifying microarchitecture, and currently understood structure-function relationships.
Collapse
Affiliation(s)
- Sara Sacher
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY
| | - Christopher J Hernandez
- Research Division, Hospital for Special Surgery, New York, NY
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY
| | - Eve Donnelly
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY
- Research Division, Hospital for Special Surgery, New York, NY
| |
Collapse
|
30
|
Comprehensive in vitro comparison of cellular and osteogenic response to alternative biomaterials for spinal implants. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112251. [PMID: 34225890 DOI: 10.1016/j.msec.2021.112251] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 02/02/2023]
Abstract
A variety of novel biomaterials are emerging as alternatives to conventional metals and alloys, for use in spinal implants. These promise potential advantages with respect to e.g. elastic modulus compatibility with the host bone, improved radiological imaging or enhanced cellular response to facilitate osseointegration. However, to date there is scarce comparative data on the biological response to many of these biomaterials that would give insights into the relative level of bone formation, resorption inhibition and inflammation. Thus, in this study, we aimed to evaluate and compare the in vitro biological response to standard discs of four alternative biomaterials: polyether ether ketone (PEEK), zirconia toughened alumina (ZTA), silicon nitride (SN) and surface-textured silicon nitride (ST-SN), and the reference titanium alloy Ti6Al4V (TI). Material-specific characteristics of these biomaterials were evaluated, such as surface roughness, wettability, protein adsorption (BSA) and apatite forming capacity in simulated body fluid. The activity of pre-osteoblasts seeded on the discs was characterized, by measuring viability, proliferation, attachment and morphology. Then, the osteogenic differentiation of pre-osteoblasts was compared in vitro from early to late stage by Alizarin Red S staining and real-time PCR analysis. Finally, osteoclast activity and inflammatory response were assessed by real-time PCR analysis. Compared to TI, all other materials generally demonstrated a lower osteoclastic activity and inflammatory response. ZTA and SN showed generally an enhanced osteogenic differentiation and actin length. Overall, we could show that SN and ST-SN showed a higher osteogenic effect than the other reference groups, an inhibitive effect against bone resorption and low inflammation, and the results indicate that silicon nitride has a promising potential to be developed further for spinal implants that require enhanced osseointegration.
Collapse
|
31
|
Tertuliano OA, Edwards BW, Meza LR, Deshpande VS, Greer JR. Nanofibril-mediated fracture resistance of bone. BIOINSPIRATION & BIOMIMETICS 2021; 16:035001. [PMID: 33470971 DOI: 10.1088/1748-3190/abdd9d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
Natural hard composites like human bone possess a combination of strength and toughness that exceeds that of their constituents and of many engineered composites. This augmentation is attributed to their complex hierarchical structure, spanning multiple length scales; in bone, characteristic dimensions range from nanoscale fibrils to microscale lamellae to mesoscale osteons and macroscale organs. The mechanical properties of bone have been studied, with the understanding that the isolated microstructure at micro- and nano-scales gives rise to superior strength compared to that of whole tissue, and the tissue possesses an amplified toughness relative to that of its nanoscale constituents. Nanoscale toughening mechanisms of bone are not adequately understood at sample dimensions that allow for isolating salient microstructural features, because of the challenge of performing fracture experiments on small-sized samples. We developed anin situthree-point bend experimental methodology that probes site-specific fracture behavior of micron-sized specimens of hard material. Using this, we quantify crack initiation and growth toughness of human trabecular bone with sharp fatigue pre-cracks and blunt notches. Our findings indicate that bone with fatigue cracks is two times tougher than that with blunt cracks.In situdata-correlated electron microscopy videos reveal this behavior arises from crack-bridging by nanoscale fibril structure. The results reveal a transition between fibril-bridging (∼1μm) and crack deflection/twist (∼500μm) as a function of length-scale, and quantitatively demonstrate hierarchy-induced toughening in a complex material. This versatile approach enables quantifying the relationship between toughness and microstructure in various complex material systems and provides direct insight for designing biomimetic composites.
Collapse
Affiliation(s)
- Ottman A Tertuliano
- Mechanical Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Bryce W Edwards
- Division of Engineering and Applied Sciences, California Institute of Technology Pasadena, CA 91125, United States of America
| | - Lucas R Meza
- Mechanical Engineering, University of Washington, Seattle, WA 98115, United States of America
| | - Vikram S Deshpande
- Department of Engineering, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Julia R Greer
- Division of Engineering and Applied Sciences, California Institute of Technology Pasadena, CA 91125, United States of America
| |
Collapse
|
32
|
Buccino F, Colombo C, Vergani LM. A Review on Multiscale Bone Damage: From the Clinical to the Research Perspective. MATERIALS (BASEL, SWITZERLAND) 2021; 14:1240. [PMID: 33807961 PMCID: PMC7962058 DOI: 10.3390/ma14051240] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/10/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023]
Abstract
The investigation of bone damage processes is a crucial point to understand the mechanisms of age-related bone fractures. In order to reduce their impact, early diagnosis is key. The intricate architecture of bone and the complexity of multiscale damage processes make fracture prediction an ambitious goal. This review, supported by a detailed analysis of bone damage physical principles, aims at presenting a critical overview of how multiscale imaging techniques could be used to implement reliable and validated numerical tools for the study and prediction of bone fractures. While macro- and meso-scale imaging find applications in clinical practice, micro- and nano-scale imaging are commonly used only for research purposes, with the objective to extract fragility indexes. Those images are used as a source for multiscale computational damage models. As an example, micro-computed tomography (micro-CT) images in combination with micro-finite element models could shed some light on the comprehension of the interaction between micro-cracks and micro-scale bone features. As future insights, the actual state of technology suggests that these models could be a potential substitute for invasive clinical practice for the prediction of age-related bone fractures. However, the translation to clinical practice requires experimental validation, which is still in progress.
Collapse
Affiliation(s)
| | | | - Laura Maria Vergani
- Department of Mechanical Engineering (DMEC), Politecnico di Milano, Via La Masa 1, 20154 Milano, Italy; (F.B.); (C.C.)
| |
Collapse
|
33
|
Zimmermann EA, Fiedler IAK, Busse B. Breaking new ground in mineralized tissue: Assessing tissue quality in clinical and laboratory studies. J Mech Behav Biomed Mater 2020; 113:104138. [PMID: 33157423 DOI: 10.1016/j.jmbbm.2020.104138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 09/15/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023]
Abstract
Mineralized tissues, such as bone and teeth, have extraordinary mechanical properties of both strength and toughness. This mechanical behavior originates from deformation and fracture resistance mechanisms in their multi-scale structure. The term quality describes the matrix composition, multi-scale structure, remodeling dynamics, water content, and micro-damage accumulation in the tissue. Aging and disease result in changes in the tissue quality that may reduce strength and toughness and lead to elevated fracture risk. Therefore, the capability to measure the quality of mineralized tissues provides critical information on disease progression and mechanical integrity. Here, we provide an overview of clinical and laboratory-based techniques to assess the quality of mineralized tissues in health and disease. Current techniques used in clinical settings include radiography-based (radiographs, dual energy x-ray absorptiometry, EOS) and x-ray tomography-based methods (high resolution peripheral quantitative computed tomography, cone beam computed tomography). In the laboratory, tissue quality can be investigated in ex vivo samples with x-ray imaging (micro and nano-computed tomography, x-ray microscopy), electron microscopy (scanning/transmission electron imaging (SEM/STEM), backscattered scanning electron microscopy, Focused Ion Beam-SEM), light microscopy, spectroscopy (Raman spectroscopy and Fourier transform infrared spectroscopy) and assessment of mechanical behavior (mechanical testing, fracture mechanics and reference point indentation). It is important for clinicians and basic science researchers to be aware of the techniques available in different types of research. While x-ray imaging techniques translated to the clinic have provided exceptional advancements in patient care, the future challenge will be to incorporate high-resolution laboratory-based bone quality measurements into clinical settings to broaden the depth of information available to clinicians during diagnostics, treatment and management of mineralized tissue pathologies.
Collapse
Affiliation(s)
| | - Imke A K Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
34
|
Quantitative and qualitative bone imaging: A review of synchrotron radiation microtomography analysis in bone research. J Mech Behav Biomed Mater 2020; 110:103887. [DOI: 10.1016/j.jmbbm.2020.103887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/13/2020] [Accepted: 05/25/2020] [Indexed: 01/07/2023]
|
35
|
Lin X, Zhao J, Gao L, Zhang C, Gao H. Ratcheting-fatigue behavior of trabecular bone under cyclic tensile-compressive loading. J Mech Behav Biomed Mater 2020; 112:104003. [PMID: 32823002 DOI: 10.1016/j.jmbbm.2020.104003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 10/23/2022]
Abstract
This study aims to investigate the ratcheting-fatigue behaviors of trabecular bone under cyclic tension-compression, which are produced due to the accumulations of residual strain in trabecular bone. Simultaneously, the effects of different loading conditions on ratcheting behaviors of trabecular bone were probed. It is found that the gap between ratcheting strains under three stress amplitudes will gradually widen. As the stress amplitude increases, the ratcheting strain also increases. Mean stress has a significant effect on the ratcheting strain. When the mean stress is 0 MPa and 0.155 MPa, the ratcheting strain increases with the number of cycles. However, when the mean stress is -0.155 MPa, the ratcheting strain decreases as the cycle goes on. The existence of double stress peak holding time causes the creep deformation of trabecular bone, which leads to the increase of ratcheting strain. It is also noted that the ratcheting strain is greatly increased with prolongation of stress peak holding time. The digital image correlation (DIC) technique was applied to analyze the fatigue failure of trabecular bone under cyclic tension-compression. It is found that the increase of stress amplitude accelerates the damage of sample and further reduces its fatigue life. Cracks are observed in trabecular bone sample, and it is noted that the crack propagation is rapid during cyclic loading.
Collapse
Affiliation(s)
- Xianglong Lin
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, PR China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, PR China
| | - Jie Zhao
- Institute of Coal Chemistry, Chinese Academy of Science, Taiyuan, 030001, PR China
| | - Lilan Gao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, PR China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, PR China.
| | - Chunqiu Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, PR China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, PR China
| | - Hong Gao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
36
|
Pendleton EG, Tehrani KF, Barrow RP, Mortensen LJ. Second harmonic generation characterization of collagen in whole bone. BIOMEDICAL OPTICS EXPRESS 2020; 11:4379-4396. [PMID: 32923050 PMCID: PMC7449751 DOI: 10.1364/boe.391866] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 05/24/2023]
Abstract
Bone is a unique biological composite material made up of a highly structured collagen mesh matrix and mineral deposits. Although mineral provides stiffness, collagen's secondary organization provides a critical role in bone elasticity. Here, we performed polarimetric analysis of bone collagen fibers using second harmonic generation (SHG) imaging to evaluate lamella sheets and collagen fiber integrity in intact cranial bone. Our polarimetric data was fitted to a model accounting for diattenuation, polarization cross-talk, and birefringence. We compared our data to the fitted model and found no significant difference between our polarimetric observation and the representation of these scattering properties up to 70 µm deep. We also observed a loss of resolution as we imaged up to 70 µm deep into bone but a conservation of polarimetric response. Polarimetric SHG allows for the discrimination of collagen lamellar sheet structures in intact bone. Our work could allow for label-free identification of disease states and monitor the efficacy of therapies for bone disorders.
Collapse
Affiliation(s)
- Emily G. Pendleton
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Kayvan F. Tehrani
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Ruth P. Barrow
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Luke J. Mortensen
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
37
|
Reply to Zadpoor: Fatigue mechanisms observed in bone provide insight to microarchitectured materials. Proc Natl Acad Sci U S A 2020; 117:6986. [PMID: 32127470 DOI: 10.1073/pnas.2000331117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
Schoeb M, Hamdy NAT, Malgo F, Winter EM, Appelman-Dijkstra NM. Added Value of Impact Microindentation in the Evaluation of Bone Fragility: A Systematic Review of the Literature. Front Endocrinol (Lausanne) 2020; 11:15. [PMID: 32117052 PMCID: PMC7020781 DOI: 10.3389/fendo.2020.00015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/09/2020] [Indexed: 12/22/2022] Open
Abstract
The current gold standard for the diagnosis of osteoporosis and the prediction of fracture risk is the measurement of bone mineral density (BMD) using dual energy x-ray absorptiometry (DXA). A low BMD is clearly associated with increased fracture risk, but BMD is not the only determinant of bone strength, particularly in secondary osteoporosis and metabolic bone disorders in which components other than BMD are affected and DXA often underestimates true fracture risk. Material properties of bone which significantly contribute to bone strength have become evaluable in vivo with the impact microindentation (IMI) technique using the OsteoProbe® device. The question arises whether this new tool is of added value in the evaluation of bone fragility. To this effect, we conducted a systematic review of all clinical studies using IMI in vivo in humans also addressing practical aspects of the technique and differences in study design, which may impact outcome. Search data generated 38 studies showing that IMI can identify patients with primary osteoporosis and fractures, patients with secondary osteoporosis due to various underlying systemic disorders, and scarce longitudinal data also show that this tool can detect changes in bone material strength index (BMSi), following bone-modifying therapy including use of corticosteroids. However, this main outcome parameter was not always concordant between studies. This systematic review also identified a number of factors that impact on BMSi outcome. These include subject- and disease-related factors such as the relationship between BMSi and age, geographical region and the presence of fractures, and technique- and operator-related factors. Taken together, findings from this systematic review confirm the added value of IMI for the evaluation and follow-up of elements of bone fragility, particularly in secondary osteoporosis. Notwithstanding, the high variability of BMSi outcome between studies calls for age-dependent reference values, and for the harmonization of study protocols. Prospective multicenter trials using standard operating procedures are required to establish the value of IMI in the prediction of future fracture risk, before this technique is introduced in routine clinical practice.
Collapse
|
39
|
Colombo C, Libonati F, Rinaudo L, Bellazzi M, Ulivieri FM, Vergani L. A new finite element based parameter to predict bone fracture. PLoS One 2019; 14:e0225905. [PMID: 31805121 PMCID: PMC6894848 DOI: 10.1371/journal.pone.0225905] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/14/2019] [Indexed: 12/18/2022] Open
Abstract
Dual Energy X-Ray Absorptiometry (DXA) is currently the most widely adopted non-invasive clinical technique to assess bone mineral density and bone mineral content in human research and represents the primary tool for the diagnosis of osteoporosis. DXA measures areal bone mineral density, BMD, which does not account for the three-dimensional structure of the vertebrae and for the distribution of bone mass. The result is that longitudinal DXA can only predict about 70% of vertebral fractures. This study proposes a complementary tool, based on Finite Element (FE) models, to improve the DXA accuracy. Bone is simulated as elastic and inhomogeneous material, with stiffness distribution derived from DXA greyscale images of density. The numerical procedure simulates a compressive load on each vertebra to evaluate the local minimum principal strain values. From these values, both the local average and the maximum strains are computed over the cross sections and along the height of the analysed bone region, to provide a parameter, named Strain Index of Bone (SIB), which could be considered as a bone fragility index. The procedure is initially validated on 33 cylindrical trabecular bone samples obtained from porcine lumbar vertebrae, experimentally tested under static compressive loading. Comparing the experimental mechanical parameters with the SIB, we could find a higher correlation of the ultimate stress, σULT, with the SIB values (R2adj = 0.63) than that observed with the conventional DXA-based clinical parameters, i.e. Bone Mineral Density, BMD (R2adj = 0.34) and Trabecular Bone Score, TBS (R2adj = -0.03). The paper finally presents a few case studies of numerical simulations carried out on human lumbar vertebrae. If our results are confirmed in prospective studies, SIB could be used-together with BMD and TBS-to improve the fracture risk assessment and support the clinical decision to assume specific drugs for metabolic bone diseases.
Collapse
Affiliation(s)
- Chiara Colombo
- Department of Mechanical Engineering, Politecnico di Milano, Milano, Italy
| | - Flavia Libonati
- Department of Mechanical Engineering, Politecnico di Milano, Milano, Italy
| | - Luca Rinaudo
- TECHNOLOGIC S.r.l. Hologic Italia, Lungo Dora Voghera, Torino, Italy
| | - Martina Bellazzi
- Department of Mechanical Engineering, Politecnico di Milano, Milano, Italy
| | - Fabio Massimo Ulivieri
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Nuclear Medicine-Bone Metabolic Unit, Milano, Italy
- * E-mail:
| | - Laura Vergani
- Department of Mechanical Engineering, Politecnico di Milano, Milano, Italy
| |
Collapse
|
40
|
BROWN HR, PELOSO JG, WERNER WCLAY, MECHOLSKY JJ, COHEN ND, VOGLER JB. A Thoroughbred racehorse with a unicortical palmar lateral condylar fracture returned to training 14 days after surgery: a hypothesis on the role of a single bone screw on crack propagation. J Equine Sci 2019; 30:7-12. [PMID: 30944541 PMCID: PMC6445752 DOI: 10.1294/jes.30.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/02/2018] [Indexed: 11/12/2022] Open
Abstract
A 2-year-old Thoroughbred racehorse had LF lameness that began post high-speed exercise and persisted for two days before the horse once again became sound. Diagnostic analgesia localized the lameness to the LF distal metacarpus, and a standing MRI identified a unicortical condylar fracture. A single 5.5 mm cortical screw was placed in lag fashion. The horse began hand walking at 14 days, racetrack jogging at 30 days, and racing at 5 months after the day of surgery. Placement of a single lag screw ahead of the tip of the crack in unicortical condylar fracture may be useful for reducing the recovery period for horses returning to training and racing.
Collapse
Affiliation(s)
| | | | | | - John J. MECHOLSKY
- Department of Materials Science and Engineering, University of Florida, FL
32611, U.S.A.
| | - Noah D. COHEN
- College of Veterinary Medicine, Texas A&M University, TX 77801,
U.S.A.
| | | |
Collapse
|
41
|
Nishiguchi A, Taguchi T. Osteoclast-Responsive, Injectable Bone of Bisphosphonated-Nanocellulose that Regulates Osteoclast/Osteoblast Activity for Bone Regeneration. Biomacromolecules 2019; 20:1385-1393. [PMID: 30768248 DOI: 10.1021/acs.biomac.8b01767] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An injectable bone may serve as a minimally invasive therapy for large orthopedic defects and osteoporosis and an alternative to allografting and surgical treatment. However, conventional bone substitutes lack the desirable biodegradability, bioresponsibility, and functionality to regulate the bone regeneration process. Here, we report an injectable, bioresponsive bone composed of bisphosphonate-modified nanocellulose (pNC) as a bone substitute for bone regeneration. Composites composed of nanofibrillated cellulose and β-tricalcium phosphate (β-TCP) mimic bone structures in which apatite reinforces collagen fibrils. Bisphosphonate groups on nanocellulose provide reversible, physical cross-linking with β-TCP, apatite formation, binding property to bone, and pH responsiveness. When the pH drops to ∼4.5, which corresponds to an osteoclast-induced pH decrease, pNC-β-TCP composite degrades and releases pNC. pNC suppresses osteoclast formation and pit formation. This osteoclast-responsive property allows for controlling the degradation rate of the composite. Moreover, the composite of pNC, α-tricalcium phosphate (α-TCP), and β-TCP enhances osteoblast differentiation. This injectable bone substitute of pNC that regulates osteoclast/osteoblast activity has enormous potential for the treatment of bone diseases and prevention of locomotive syndrome.
Collapse
Affiliation(s)
- Akihiro Nishiguchi
- Biomaterials Field, Research Center for Functional Materials , National Institute for Materials Science , 1-1 Namiki , Tsukuba , Ibaraki 305-0044 , Japan
| | - Tetsushi Taguchi
- Biomaterials Field, Research Center for Functional Materials , National Institute for Materials Science , 1-1 Namiki , Tsukuba , Ibaraki 305-0044 , Japan
| |
Collapse
|
42
|
Fourier Transform Infrared Spectroscopy of Bone Tissue: Bone Quality Assessment in Preclinical and Clinical Applications of Osteoporosis and Fragility Fracture. Clin Rev Bone Miner Metab 2019. [DOI: 10.1007/s12018-018-9255-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
43
|
Non-setting, injectable biomaterials containing particulate hydroxyapatite can increase primary stability of bone screws in cancellous bone. Clin Biomech (Bristol, Avon) 2018; 59:174-180. [PMID: 30268995 DOI: 10.1016/j.clinbiomech.2018.09.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 09/10/2018] [Accepted: 09/21/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Fracture fixation in weak bone is still a clinical challenge. Screw augmentation was shown to successfully increase their primary stability. The currently used calcium phosphate or polymeric bone cements, however, present important drawbacks such as induced toxicity and/or impaired bone neo-formation. A new approach to enhance bone screw primary stability without affecting bone formation is the use of non-setting, calcium phosphate loaded soft materials as the augmentation material. METHODS Two types of biomaterials (non-crosslinked hyaluronic acid as viscous fluid and agar as hydrogel) were loaded with 40 wt/vol% of hydroxyapatite particles and characterized. The screw augmentation effect of all materials was evaluated through pull-out tests in bovine cancellous bone and compared to the non-augmented situation (control). The bone mineral density of each test sample was measured with μCT scans and was used to normalize the pull-out strength. FINDINGS Both materials loaded with hydroxyapatite increased the normalized pull-out strength of the screws compared to control samples and particle-free materials. This counter-intuitive augmentation effect increased with decreasing bone mineral density and was independent from the type of the soft materials used. INTERPRETATION We were able to demonstrate that non-setting, injectable biomaterials loaded with ceramic particles can significantly enhance the primary stability of bone screws. This material combination opens the unique possibility to achieve a screw augmentation effect without impairing or even potentially favoring the bone formation in proximity to the screw. This effect would be particularly advantageous for the treatment of osteoporotic bone fractures requiring a stabilization with bone screws.
Collapse
|
44
|
He Y, Mu C, Shen X, Yuan Z, Liu J, Chen W, Lin C, Tao B, Liu B, Cai K. Peptide LL-37 coating on micro-structured titanium implants to facilitate bone formation in vivo via mesenchymal stem cell recruitment. Acta Biomater 2018; 80:412-424. [PMID: 30266635 DOI: 10.1016/j.actbio.2018.09.036] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/02/2018] [Accepted: 09/24/2018] [Indexed: 01/06/2023]
Abstract
Titanium (Ti) and Ti-alloys were widely used in clinic orthopedics, however, the insufficient bone formation surrounding Ti-based implants still limited their biological performances. Surface modification of Ti substrates is essential to improve their interactions with bone-forming cells and bone tissue. In this study, we modified Ti substrates by coating peptide LL-37 onto micro-structured Ti substrates and aimed to (i) induce mesenchymal stem cells (MSCs) migration both in vitro and in vivo, (ii) facilitate osteogenic differentiation of MSCs and new bone formation. The surface micro-structured Ti substrates with hydroxyapatite deposition were fabricated by a two-step method including micro-arc oxidation (MAO) and hydrothermal treatment. LL-37 was loaded on micro-structured Ti substrates with the assistance of polydopamine coating. We confirmed that surface-modified Ti substrates benefited viability, adhesion, migration and osteogenic differentiation of MSCs in vitro. In a femur-defect rat model, the surface-modified Ti implants effectively induced CD29+/CD90+ positive cells migration in one week after implantation. According to the results of H&E, Masson's trichrome staining and immunohistochemical staining of OCN, OPN and collagen I, the targeted Ti implants exhibited significant new bone formation after implantation for 4 weeks. These results indicate that the surface modification of Ti samples facilitated bone formation through MSCs recruitment. STATEMENT OF SIGNIFICANCE: The inherent surface bioinertness of titanium (Ti) and Ti-alloys still limits their biological performances in clinical applications. Recently, the strategy of mesenchymal stem cells (MSCs) recruitment has been proposed to improve the osteointegration of bone implants. Herein, we reports the surface modification of Ti implants from the point of MSCs recruitment. Peptide LL-37 was coated on micro-structured Ti substrates to (i) recruit MSCs, (ii) regulate bio-physiological performance of MSCs, and (iii) facilitate bone formation in vivo. Our results improve the understanding of the interaction between Ti implants and MSCs, and provide a promising strategy of MSCs recruitment in the design of bone repair related biomaterials.
Collapse
Affiliation(s)
- Ye He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Caiyun Mu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Xinkun Shen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China; School of Life Science, Chongqing University, Chongqing 400044, PR China
| | - Zhang Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Ju Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Weizhen Chen
- First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, Hangzhou, Zhejiang 310003, PR China
| | - Chuanchuan Lin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Bailong Tao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China
| | - Bin Liu
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, School of Life Science, Southwest University, Chongqing 400715, PR China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, PR China.
| |
Collapse
|
45
|
Bartlow CM, Mann KA, Damron TA, Oest ME. Limited field radiation therapy results in decreased bone fracture toughness in a murine model. PLoS One 2018; 13:e0204928. [PMID: 30281657 PMCID: PMC6169919 DOI: 10.1371/journal.pone.0204928] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/17/2018] [Indexed: 11/19/2022] Open
Abstract
Fragility fractures are a well-known complication following oncologic radiotherapy, and it is suspected that radiation-induced embrittlement of bone within the treatment field may contribute to fracture risk. To explore this phenomenon, a mouse model (BALB/cJ) of fractionated, limited field, bilateral hindlimb irradiation (4x5 Gy) was used. The effects of radiation on femoral (cortical) bone fracture toughness, morphology, and biochemistry-including advanced glycation end products (AGEs)-were quantified and compared to Sham group samples prior to irradiation and at 0, 4, 8, and 12 weeks post-irradiation. Additionally, alterations to bone fracture toughness mediated directly by radiation (independent of cellular mechanisms) were determined using devitalized mouse cadaver femurs. Finally, the contribution of AGEs to reduced fracture toughness was examined by artificially ribosylating mouse femurs ex vivo. These data demonstrate that in vivo irradiation results in an immediate (-42% at 0 weeks, p < 0.001) and sustained (-28% at 12 weeks, p < 0.001) decrease in fracture toughness with small changes in morphology (-5% in cortical area at 12 weeks), and minimal changes in bone composition (tissue mineral density, mineral:matrix ratio, and AGE content). Irradiation of devitalized femurs also reduced fracture toughness (-29%, p < 0.001), but to a lesser extent than was seen in vivo. While artificial ribosylation decreased fracture toughness with time, the extent of glycation needed to induce this effect exceeded the AGE accumulation that occurred in vivo. Overall, hindlimb irradiation induced a substantial and sustained decrease in bone fracture toughness. Approximately half of this decrease in fracture toughness is due to direct radiation damage, independent of cellular remodeling. Collagen glycation in vivo was not substantially altered, suggesting other matrix changes may contribute to post-radiotherapy bone embrittlement.
Collapse
Affiliation(s)
- Christopher M. Bartlow
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Kenneth A. Mann
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Timothy A. Damron
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Megan E. Oest
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| |
Collapse
|