1
|
Bharadwaj S, Lima F, Pathak TB, Dhamdhere R, Fu P, Malluche H, Rao M, Madabhushi A. ADAM: automated digital phenotyping and morphological texture analysis of bone biopsy images using deep learning. JBMR Plus 2025; 9:ziaf028. [PMID: 40129969 PMCID: PMC11931614 DOI: 10.1093/jbmrpl/ziaf028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 03/26/2025] Open
Abstract
Histomorphometric analysis of undecalcified bone biopsy images provides quantitative assessment of bone turnover, volume, and mineralization using static and dynamic parameters. Traditionally, quantification has relied on manual annotation and tracing of relevant tissue structures, a process that is time-intensive and subject to inter-operator variability. We developed ADAM, an automated pipeline for digital phenotyping, to quantify tissue and cellular components pertinent to static histomorphometric parameters such as bone and osteoid area, osteoclast and osteoblast count, and bone marrow adipose tissue (BMAT) area. The pipeline allowed rapid generation of delineated tissue and cell maps for up to 20 images in less than a minute. Comparing deep learning-generated annotation pixels with manual annotations, we observed Spearman correlation coefficients of ρ = 0.99 for both mineralized bone and osteoid, and ρ = 0.94 for BMAT. For osteoclast and osteoblast cell counts, which are subject to morphologic heterogeneity, using only brightfield microscopic images and without additional staining, we noted ρ = 0.60 and 0.69, respectively (inter-operator correlation was ρ = 0.62 for osteoclast and 0.84 for osteoblast count). The study also explored the application of morphological texture analysis (MTA), measuring relative pixel patterns that potentially vary with diverse tissue conditions. Notably, MTA from mineralized bone, osteoid, and BMAT showed differentiating potential to identify common pixel characteristics between images labeled as low or high bone turnover based upon the final diagnostic report of the bone biopsy. The Area Under the Receiver Operating Characteristic Curve (AUC-ROC) obtained for BMAT MTA features as a classifier for bone turnover, was 0.87, suggesting that computer-extracted features, not discernable to the human eye, hold potential in classifying tissue states. With additional evaluation, ADAM could be potentially integrated into existing clinical routines to improve pathology workflows and contribute to diagnostic insights into bone biopsy evaluation and reporting.
Collapse
Affiliation(s)
- Satvika Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
| | - Florence Lima
- Division of Nephrology, Bone and Mineral Metabolism, University of Kentucky, Lexington, KY 40508, United States
| | - Tilak Bahadur Pathak
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
| | - Rohan Dhamdhere
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Hartmut Malluche
- Division of Nephrology, Bone and Mineral Metabolism, University of Kentucky, Lexington, KY 40508, United States
| | - Madhumathi Rao
- Division of Nephrology, Bone and Mineral Metabolism, University of Kentucky, Lexington, KY 40508, United States
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
- Atlanta Veterans Affairs Medical Center, Atlanta, GA 30033, United States
| |
Collapse
|
2
|
Saiz AM, Rahmati M, Gresham RCH, Baldini TD, Burgan J, Lee MA, Osipov B, Christiansen BA, Khassawna TE, Wieland DCF, Marinho AL, Blanchet C, Czachor M, Working ZM, Bahney CS, Leach JK. Polytrauma impairs fracture healing accompanied by increased persistence of innate inflammatory stimuli and reduced adaptive response. J Orthop Res 2025; 43:603-616. [PMID: 39550711 PMCID: PMC11806648 DOI: 10.1002/jor.26015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/23/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
The field of bone regeneration has primarily focused on investigating fracture healing and nonunion in isolated musculoskeletal injuries. Compared to isolated fractures, which frequently heal well, fractures in patients with multiple bodily injuries (polytrauma) may exhibit impaired healing. While some papers have reported the overall cytokine response to polytrauma conditions, significant gaps in our understanding remain in how fractures heal differently in polytrauma patients. We aimed to characterize fracture healing and the temporal local and systemic immune responses to polytrauma in a murine model of polytrauma composed of a femur fracture combined with isolated chest trauma. We collected serum, bone marrow from the uninjured limb, femur fracture tissue, and lung tissue over 3 weeks to study the local and systemic immune responses and cytokine expression after injury. Immune cell distribution was assessed by flow cytometry. Fracture healing was characterized using microcomputed tomography (microCT), histological staining, immunohistochemistry, mechanical testing, and small angle X-ray scattering. We detected more innate immune cells in the polytrauma group, both locally at the fracture site and systemically, compared to other groups. The percentage of B and T cells was dramatically reduced in the polytrauma group 6 h after injury and remained low throughout the study duration. Fracture healing in the polytrauma group was impaired, evidenced by the formation of a poorly mineralized and dysregulated fracture callus. Our data confirm the early, dysregulated inflammatory state in polytrauma that correlates with disorganized and impaired fracture healing.
Collapse
Affiliation(s)
| | - Maryam Rahmati
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| | | | - Tony Daniel Baldini
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
- California Northstate University College of MedicineSacramentoCaliforniaUSA
| | - Jane Burgan
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
- Stony Brook Renaissance School of MedicineStony BrookNew YorkUSA
| | - Mark A. Lee
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| | - Benjamin Osipov
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| | | | - Thaqif El Khassawna
- Experimental Trauma SurgeryJustus‐Liebig University GiessenGiessenGermany
- Faculty of Health SciencesUniversity of Applied SciencesGiessenGermany
| | | | - André Lopes Marinho
- Institute of Metallic Biomaterials, Helmholtz Zentrum HereonGeesthachtGermany
| | | | - Molly Czachor
- Steadman Phillippon Research InstituteVailColoradoUSA
| | | | - Chelsea S. Bahney
- Steadman Phillippon Research InstituteVailColoradoUSA
- University of CaliforniaSan FranciscoCaliforniaUSA
| | - J. Kent Leach
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| |
Collapse
|
3
|
Kang MH, Kim HB, Chung JH, Choung PH. Parameter-Tuned Pulsed Wave Photobiomodulation Enhances Stem Cells From Apical Papilla Differentiation: Evidence From Gene and Protein Analyses. JOURNAL OF BIOPHOTONICS 2025; 18:e202400348. [PMID: 39807665 DOI: 10.1002/jbio.202400348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025]
Abstract
This study examines the effects of pulsed wave photobiomodulation (pwPBM) on the osteogenic differentiation of stem cells from the apical papilla (SCAP). Using 810 nm near-infrared (NIR) light with 300 Hz pulses and a 30% duty cycle, pwPBM was applied at a total energy density of 750 mJ/cm2. Osteogenesis was evaluated through both in vitro and in vivo analyses. In vitro experiments demonstrated significant enhancement of alkaline phosphatase (ALP) activity, along with upregulation of key osteogenesis-related genes and proteins, as confirmed by real-time polymerase chain reaction (PCR) and Western blot analyses. In vivo, histological assessments following SCAP transplantation revealed increased bone tissue formation, further corroborated by osteocalcin staining. These findings underscore the potential of pwPBM as an innovative and effective tool for dental tissue regeneration and engineering.
Collapse
Affiliation(s)
- Moon-Ho Kang
- Department of Oral and Maxillofacial Surgery and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Hong Bae Kim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, Republic of Korea
- BioEngineering Institute of Chang Healthcare, co. Ltd, Seoul, Republic of Korea
| | - Jong Hoon Chung
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, Republic of Korea
| | - Pill-Hoon Choung
- Department of Oral and Maxillofacial Surgery and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Genoud KJ, Sadowska JM, Power RN, Costard LS, Ryan EJ, Matherson AR, Gonzalez-Vazquez AG, Lemoine M, Eichholz K, Pitacco P, Chen G, Cavanagh B, Garcia O, Murphy CM, Curtin CM, Kelly DJ, O'Brien FJ. Collagen silver-doped hydroxyapatite scaffolds reinforced with 3D printed frameworks for infection prevention and enhanced repair of load-bearing bone defects. Biofabrication 2025; 17:025010. [PMID: 39874655 DOI: 10.1088/1758-5090/adaf59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/28/2025] [Indexed: 01/30/2025]
Abstract
Osteomyelitis, a severe bone infection, is an extremely challenging complication in the repair of traumatic bone defects. Furthermore, the use of long-term high-dose antibiotics in standard treatment increases the risks of antibiotic resistance. Herein, an antibiotic-free, collagen silver-doped hydroxyapatite (coll-AgHA) scaffold reinforced with a 3D printed polycaprolactone (PCL) framework was developed with enhanced mechanical properties to be used in the repair of load-bearing defects with antimicrobial properties as a preventative measure against osteomyelitis. The AgHA particles were fabricated in varying Ag doses and loaded within freeze-dried collagen scaffolds at two concentrations. The optimised Ag dose (1.5 mol% Ag) and AgHA concentration (200 wt%) within the collagen scaffold demonstratedin vitroosteogenic and antibacterial properties againstS. aureus (S. aureus),the main causative pathogen of osteomyelitis. The addition of the PCL framework to the coll-AgHA scaffolds significantly enhanced the compressive modulus from 4 to 12 MPa while maintaining high porosity as well as both pro-osteogenic and antibacterial properties. The reinforced coll-AgHA scaffolds were implantedin vivoand demonstrated enhanced bone repair, significantly greater vessel formation, and calcified tissue in a load-bearing critical sized defect in rats. Taken together, these results confirm the capacity of this novel biomaterial scaffold as a preventative measure against infection in bone repair for use in load-bearing defects, without the use of antibiotics.
Collapse
Affiliation(s)
- Katelyn J Genoud
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Joanna M Sadowska
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Rachael N Power
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Lara S Costard
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Emily J Ryan
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Austyn R Matherson
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Arlyng G Gonzalez-Vazquez
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Mark Lemoine
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Kian Eichholz
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Pierluca Pitacco
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Gang Chen
- Microsurgical Research and Training Facility (MRTF), RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Orquidea Garcia
- Johnson & Johnson 3D Printing Innovation & Customer Solutions, Johnson & Johnson Services, Inc., Irvine, CA, United States of America
| | - Ciara M Murphy
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Daniel J Kelly
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Biomedical Engineering Research (AMBER) Centre, Dublin, Ireland
| |
Collapse
|
5
|
Shah MAA, Tang W, Zhang JH, Chen C, Wang JW, Lü SJ, Yu XT, Zhang ZJ, Li C, Yu SB, Sui HJ. Microvasculature and trabecular bone in beagle proximal femur: Microstructural insights. Ann Anat 2025; 258:152368. [PMID: 39643064 DOI: 10.1016/j.aanat.2024.152368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Avascular necrosis of femoral head and malunion are frequent post-operative complications of femoral neck fractures. To optimize surgical techniques, this study aims to provide a microstructural understanding of intraosseous microvasculature and the trabecular bone of the femoral head and neck. STUDY DESIGN This anatomical study analyzed twenty-eight femora from fourteen cadaveric beagles. Common iliac arteries were infused with colored silicone-resin for vascular visualization, followed by non-decalcified hard tissue processing using the EXAKT®, and Masson's trichrome staining. Morphology and histomorphometric analysis were performed by Nikon NIS Elements BR and ImageJ-fiji. RESULTS Histomorphometry revealed thin, elongated trabeculae with high vascularity aligned parallel in the neck; numerous intraosseous anastomoses at the neck-shaft and head-neck junctions; thick trabeculae with smaller marrow cavities, and dense branching vascular networks near the cortex in the head. Quantitative analysis showed an inverse correlation between trabecular density and mean vascular density/vascular length density, with no significant sex or side differences. Dense connective tissue fibers maintained the microvasculature and trabeculae structure. CONCLUSION The femoral neck displayed an outside-in microvascular pattern via retinacular branches. Conversely, the femoral head had an inside-out pattern through epiphyseal branches reinforced by medullary branches. Dense intraosseous microvasculature aligned sub-cortically. The study identified a potential anatomical safe zone for screwing in femoral neck fractures in beagles. These findings provide an anatomical basis for translational research in joint preservation techniques for humans.
Collapse
Affiliation(s)
- M Adeel Alam Shah
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China.
| | - Wei Tang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Jing-Hui Zhang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Cheng Chen
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Jia-Wei Wang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Shu-Jun Lü
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Xin-Tong Yu
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Zhi-Jun Zhang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Chan Li
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Sheng-Bo Yu
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China.
| | - Hong-Jin Sui
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
6
|
Wang Z, Kregel M, Meijers JL, Franch J, Cuijpers VMJI, Ahlers D, Karst U, Slootweg P, van der Geest IC, Leeuwenburgh SC, van den Beucken JJ. Cisplatin-functionalized dual-functional bone substitute granules for bone defect treatment after bone tumor resection. Acta Biomater 2025; 191:158-176. [PMID: 39551330 DOI: 10.1016/j.actbio.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Invasive bone tumors pose a significant healthcare challenge, often requiring systemic chemotherapy and limb salvage surgery. However, these strategies are hampered by severe side effects, complex post-resection bone defects, and high local recurrence rates. To address this, we developed dual-functional bone substitute biomaterials by functionalizing commercially available bone substitute granules (Bio-Oss® and MBCP®+) with the established anticancer agent cisplatin. Physicochemical characterization revealed that Bio-Oss® granules possess a higher surface area and lower crystallinity compared to MBCP®+ granules, which enhances their capacity for cisplatin adsorption and release. In co-cultures with metastatic breast and prostate cancer cells (MDA-MB-231 and PC3) and bone marrow stromal cells (hBMSCs), cisplatin-functionalized granules and their releasates exhibited dose-dependent cytotoxic effects on cancer cells while having less impact on hBMSCs. Furthermore, investigations on the mechanism of action indicated that cisplatin induced significant cell cycle arrest and apoptosis in MDA-MB-231 and PC3 cells, contrasting with minimal effects on hBMSCs. In a rat femoral condyle defect model, cisplatin-functionalized granules did not evoke adverse effects on bone tissue ingrowth or new bone formation. Importantly, local application of cisplatin-functionalized granules resulted in negligible cisplatin accumulation without signs of apoptotic damage in kidneys and livers. Taken together, we here provide hard evidence that cisplatin-functionalized granules maintain a favorable balance between biosafety, anticancer efficacy, and bone regenerative capacity. Consequently, loading granular bone substitutes with cisplatin holds promise for local treatment of bone defects following bone tumor resections, presenting a safe and potentially more effective alternative to systemic cisplatin administration. STATEMENT OF SIGNIFICANCE: Current treatments in combating malignant bone tumors are hampered by severe side effects, high local tumor recurrence, and complex bone defects after surgery. This study explores a facile manufacturing method to render two types of commercially available bone substitute granules (Bio-Oss® and MBCP®+) suitable for local delivery of cisplatin. The use of cisplatin-functionalized granules has shown promising results both in killing cancer cells in a dose-dependent manner and in aiding bone regeneration. Importantly, this local treatment strategy avoids the systemic toxicity associated with traditional chemotherapy to excretory organs. This dual-functional strategy represents a significant advancement in bone cancer treatment, offering a safe and more efficient alternative that could improve outcomes for patients following bone tumor resection.
Collapse
Affiliation(s)
- Zhule Wang
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Mark Kregel
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jean-Luc Meijers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jordi Franch
- Department of Small Animal Medicine and Surgery, Veterinary School, Universitat Autonoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Vincent M J I Cuijpers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - David Ahlers
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Piet Slootweg
- Department of Pathology, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Ingrid Cm van der Geest
- Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands; Department of Orthopedics, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Sander Cg Leeuwenburgh
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Jeroen Jjp van den Beucken
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands.
| |
Collapse
|
7
|
Rogova VV, Peev S, Yotsova R, Gerova-Vatsova T, Parushev I. Histomorphometric Assessment of Non-Decalcified Plastic-Embedded Specimens for Evaluation of Bone Regeneration Using Bone Substitute Materials-A Systematic Review. MATERIALS (BASEL, SWITZERLAND) 2024; 18:119. [PMID: 39795764 PMCID: PMC11722015 DOI: 10.3390/ma18010119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025]
Abstract
With the implementation of bone substitute materials, regeneration strategies have inevitably evolved over the years. Histomorphometry is the optimal means of quantitative evaluation of bone structure and morphology. This systematic review focuses on determining study models, staining methods and histomorphometric parameters used for bone regeneration research on non-decalcified plastic-embedded specimens over the last 10 years. After being subjected to the inclusion and exclusion criteria, 118 studies were included in this review. The results establish the most commonly selected animal model is rat, followed by rabbit, sheep and dog. Strong preference for staining samples with toluidine blue was noted. With regard to histomorphometric parameters, terms related to bone were most frequently assessed, amounting to almost half of recorded parameters. New bone formation was the main descriptor of this category. Residual bone graft and non-bone tissue parameters were also often evaluated. With regard to dynamic histomorphometry, mineral apposition rate (MAR) was the parameter of choice for most researchers, with calcein green being the preferred dye for fluorochrome labelling. An overview of the contemporary literature, as well as weaknesses in the current research protocols have been discussed.
Collapse
Affiliation(s)
- Varvara-Velika Rogova
- Department of Oral Surgery, Faculty of Dental Medicine, Medical University of Varna, 9002 Varna, Bulgaria;
| | - Stefan Peev
- Department of Periodontology and Dental Implantology, Faculty of Dental Medicine, Medical University of Varna, 9002 Varna, Bulgaria; (S.P.); (T.G.-V.)
| | - Ralitsa Yotsova
- Department of Oral Surgery, Faculty of Dental Medicine, Medical University of Varna, 9002 Varna, Bulgaria;
| | - Tsvetalina Gerova-Vatsova
- Department of Periodontology and Dental Implantology, Faculty of Dental Medicine, Medical University of Varna, 9002 Varna, Bulgaria; (S.P.); (T.G.-V.)
| | - Ivaylo Parushev
- Department of Clinical Medical Sciences, Faculty of Dental Medicine, Medical University of Varna, 9002 Varna, Bulgaria;
| |
Collapse
|
8
|
Pereira KA, Torquato LC, Maciel CCM, Nunes CMM, Mantovani LO, Almeida ND, Lopes SLPC, de Vasconcellos LMR, Jardini MAN, Marcuzzo JS, De Marco AC. Carbon fiber felt scaffold from Brazilian textile PAN fiber for regeneration of critical size bone defects in rats: A histomorphometric and microCT study. J Biomed Mater Res B Appl Biomater 2024; 112:e35467. [PMID: 39180195 DOI: 10.1002/jbm.b.35467] [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: 02/12/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
The objective of the present study was to evaluate the carbon fiber obtained from textile PAN fiber, in its different forms, as a potential scaffolds synthetic bone. Thirty-four adult rats were used (Rattus norvegicus, albinus variation), two critical sized bone defects were made that were 5 mm in diameter. Twenty-four animals were randomly divided into four groups: control (C)-bone defect + blood clot, non-activated carbon fiber felt (NACFF)-bone defect + NACFF, activated carbon fiber felt (ACFF)-bone defect + ACFF, and silver activated carbon fiber felt (Ag-ACFF)-bone defect + Ag-ACFF, and was observed by 15 and 60 days for histomorphometric, three-dimensional computerized microtomography (microCT) and mineral apposition analysis. On histomorphometric and microCT analyses, NACFF were associated with higher proportion of neoformed bone and maintenance of bone structure. On fluorochrome bone label, there was no differences between the groups. NACFF has shown to be a promising synthetic material as a scaffold for bone regeneration.
Collapse
Affiliation(s)
- Kauê Alberto Pereira
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), Institute of Science and Technology, São Jose dos Campos, São Paulo, Brazil
- Division of Periodontology, Fundação Universitária Vida Cristã, Pindamonhangaba, São Paulo, Brazil
| | - Letícia Cavassini Torquato
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), Institute of Science and Technology, São Jose dos Campos, São Paulo, Brazil
| | - Clarissa Carvalho Martins Maciel
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), Institute of Science and Technology, São Jose dos Campos, São Paulo, Brazil
| | - Camilla Magnoni Moretto Nunes
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), Institute of Science and Technology, São Jose dos Campos, São Paulo, Brazil
| | - Ludmilla Oliveira Mantovani
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), Institute of Science and Technology, São Jose dos Campos, São Paulo, Brazil
| | - Nátaly Domingues Almeida
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), Institute of Science and Technology, São Jose dos Campos, São Paulo, Brazil
| | - Sergio Lucio Pereira Castro Lopes
- Department of Diagnosis and Surgery-Division of Radiology, São Paulo State University (UNESP), Institute of Science and Technology, São José dos Campos, Brazil
| | - Luana Marotta Reis de Vasconcellos
- Department of Biosciences and Oral Diagnosis-Division of Histology, São Paulo State University (UNESP), Institute of Science and Technology, São José dos Campos, Brazil
| | - Maria Aparecida Neves Jardini
- Department of Diagnosis and Surgery-Division of Periodontology, São Paulo State University (UNESP), Institute of Science and Technology, São José dos Campos, Brazil
| | | | - Andrea Carvalho De Marco
- Department of Diagnosis and Surgery-Division of Periodontology, São Paulo State University (UNESP), Institute of Science and Technology, São José dos Campos, Brazil
| |
Collapse
|
9
|
Nelson AL, Mancino C, Gao X, Choe JA, Chubb L, Williams K, Czachor M, Marcucio R, Taraballi F, Cooke JP, Huard J, Bahney C, Ehrhart N. β-catenin mRNA encapsulated in SM-102 lipid nanoparticles enhances bone formation in a murine tibia fracture repair model. Bioact Mater 2024; 39:273-286. [PMID: 38832305 PMCID: PMC11145078 DOI: 10.1016/j.bioactmat.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024] Open
Abstract
Fractures continue to be a global economic burden as there are currently no osteoanabolic drugs approved to accelerate fracture healing. In this study, we aimed to develop an osteoanabolic therapy which activates the Wnt/β-catenin pathway, a molecular driver of endochondral ossification. We hypothesize that using an mRNA-based therapeutic encoding β-catenin could promote cartilage to bone transformation formation by activating the canonical Wnt signaling pathway in chondrocytes. To optimize a delivery platform built on recent advancements in liposomal technologies, two FDA-approved ionizable phospholipids, DLin-MC3-DMA (MC3) and SM-102, were used to fabricate unique ionizable lipid nanoparticle (LNP) formulations and then tested for transfection efficacy both in vitro and in a murine tibia fracture model. Using firefly luciferase mRNA as a reporter gene to track and quantify transfection, SM-102 LNPs showed enhanced transfection efficacy in vitro and prolonged transfection, minimal fracture interference and no localized inflammatory response in vivo over MC3 LNPs. The generated β-cateninGOF mRNA encapsulated in SM-102 LNPs (SM-102-β-cateninGOF mRNA) showed bioactivity in vitro through upregulation of downstream canonical Wnt genes, axin2 and runx2. When testing SM-102-β-cateninGOF mRNA therapeutic in a murine tibia fracture model, histomorphometric analysis showed increased bone and decreased cartilage composition with the 45 μg concentration at 2 weeks post-fracture. μCT testing confirmed that SM-102-β-cateninGOF mRNA promoted bone formation in vivo, revealing significantly more bone volume over total volume in the 45 μg group. Thus, we generated a novel mRNA-based therapeutic encoding a β-catenin mRNA and optimized an SM-102-based LNP to maximize transfection efficacy with a localized delivery.
Collapse
Affiliation(s)
- Anna Laura Nelson
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
- Colorado State University, School of Biomedical Engineering, Fort Collins CO, USA
| | - Chiara Mancino
- Houston Methodist Research Institute, Center for Musculoskeletal Regeneration, Houston TX, USA
| | - Xueqin Gao
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
| | - Joshua A. Choe
- University of Wisconsin-Madison, Department of Orthopedics and Rehabilitation, Department of Biomedical Engineering, Medical Scientist Training Program, Madison, WI, USA
| | - Laura Chubb
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
| | - Katherine Williams
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Molly Czachor
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
| | - Ralph Marcucio
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - Francesca Taraballi
- Houston Methodist Research Institute, Center for Musculoskeletal Regeneration, Houston TX, USA
| | - John P. Cooke
- Houston Methodist Research Institute, Center for RNA Therapeutics, Department of Cardiovascular Sciences, Houston, TX, USA
| | - Johnny Huard
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
| | - Chelsea Bahney
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - Nicole Ehrhart
- Colorado State University, School of Biomedical Engineering, Fort Collins CO, USA
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| |
Collapse
|
10
|
Zainal Ariffin SH, Megat Abdul Wahab R, Abdul Razak M, Yazid MD, Shahidan MA, Miskon A, Zainol Abidin IZ. Evaluation of in vitro osteoblast and osteoclast differentiation from stem cell: a systematic review of morphological assays and staining techniques. PeerJ 2024; 12:e17790. [PMID: 39071131 PMCID: PMC11283775 DOI: 10.7717/peerj.17790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Background Understanding human stem cell differentiation into osteoblasts and osteoclasts is crucial for bone regeneration and disease modeling. Numerous morphological techniques have been employed to assess this differentiation, but a comprehensive review of their application and effectiveness is lacking. Methods Guided by the PRISMA framework, we conducted a rigorous search through the PubMed, Web of Science and Scopus databases, analyzing 254 articles. Each article was scrutinized against pre-defined inclusion criteria, yielding a refined selection of 14 studies worthy of in-depth analysis. Results The trends in using morphological approaches were identified for analyzing osteoblast and osteoclast differentiation. The three most used techniques for osteoblasts were Alizarin Red S (mineralization; six articles), von Kossa (mineralization; three articles) and alkaline phosphatase (ALP; two articles) followed by one article on Giemsa staining (cell morphology) and finally immunochemistry (three articles involved Vinculin, F-actin and Col1 biomarkers). For osteoclasts, tartrate-resistant acid phosphatase (TRAP staining) has the highest number of articles (six articles), followed by two articles on DAPI staining (cell morphology), and immunochemistry (two articles with VNR, Cathepsin K and TROP2. The study involved four stem cell types: peripheral blood monocyte, mesenchymal, dental pulp, and periodontal ligament. Conclusion This review offers a valuable resource for researchers, with Alizarin Red S and TRAP staining being the most utilized morphological procedures for osteoblasts and osteoclasts, respectively. This understanding provides a foundation for future research in this rapidly changing field.
Collapse
Affiliation(s)
- Shahrul Hisham Zainal Ariffin
- Department of Science Biology and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Rohaya Megat Abdul Wahab
- Centre of Family Dental Health, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Muhammad Abdul Razak
- Board of Director Office, 6th Floor, Chancellery Building, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Muhammad Ashraf Shahidan
- Department of Science Biology and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Azizi Miskon
- Department of Electrical and Electronics Engineering, Faculty of Engineering, National Defence University of Malaysia, Sungai Besi, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Intan Zarina Zainol Abidin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Cyberjaya, Cyberjaya, Selangor, Malaysia
| |
Collapse
|
11
|
Xu H, Sun Z, Wang G, Li R. The Impact of Depression on Detrimental Changes in Bone Microstructure in Female Mice. Neuropsychiatr Dis Treat 2024; 20:1421-1433. [PMID: 39049938 PMCID: PMC11268775 DOI: 10.2147/ndt.s454865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
Background Several clinical studies have examined the connection between depression and bone loss, but the cause-and-effect relationship between the two conditions, especially in animal models, is not well-studied. Methods A total of 32 female mice were, randomly divided into control group (CON, n=19) and depression group (DEP, n=13). The mice in the DEP group were subjected to 21 consecutive days of restraint stress, following depressive-like behaviors were assessment. The femurs were collected using Micro-Computed Tomography (μCT) and histochemical staining. In parallel, levels of serotonin-related proteins in the brain were measured using Western blot analysis, and sex hormone profiles were determined through liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). Results The mice in the DEP group exhibited clear signs of depressive-like behaviors and an increase in serotonin transporter levels (t=-2.435, P< 0.05). In comparison to the CON mice, the DEP mice showed a decrease in bone mineral density (t =3.741, P< 0.05), bone surface area density (t =8.009, P<0.01), percent bone volume (t =4.293, P< 0.05), trabecular number (t =5.844, P<0.01), and connected density (t =11.000, P< 0.05). Additionally, there was an increase in trabecular separation (t =-7.436, P<0.01) in DEP mice. Furthermore, the DEP mice displayed a significant reduction in serum estrogen levels (t =4.340, P< 0.05) and changes in its metabolite (t =-3.325, P< 0.05), while the levels of androgens remained unchanged. Conclusion The restraint stress not only led to the development of depressive-like behaviors but also disrupted the estrogen metabolism pathway, resulting in damage to bone mass and microstructure in female mice. These findings suggest that stress-induced depression may pose a risk for bone loss in female mice by altering estrogen metabolism pathways.
Collapse
Affiliation(s)
- Hong Xu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Gang Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
12
|
Duke VR, Philippon MJ, Lind DRG, Kasler H, Yamaura K, Huard M, Czachor M, Hollenbeck J, Brown J, Garcia A, Fukase N, Marcucio RS, Nelson AL, Hambright WS, Snapper DM, Huard J, Bahney CS. Murine Progeria Model Exhibits Delayed Fracture Healing with Dysregulated Local Immune Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596277. [PMID: 38854043 PMCID: PMC11160782 DOI: 10.1101/2024.05.29.596277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Bone fracture is one of the most globally prevalent injuries, with an estimated 189 million bone fractures occurring annually. Delayed union or nonunion occurs in up to 15% of fractures and involves the interruption or complete failure of bone continuity following fracture. Preclinical testing is essential to support the translation of novel strategies to promote improved fracture repair treatment, but there is a paucity of small animal models that recapitulate clinical attributes associated with delayed fracture healing. This study explores whether the Zmpste24 -/- (Z24 -/- ) knockout mouse model of Hutchinson-Gilford progeria syndrome presents with delayed fracture healing. Leveraging the previously characterized Z24 -/- phenotype of genomic instability, epigenetic changes, and fragility, we hypothesize that these underlying alterations will lead to significantly delayed fracture healing relative to age-matched wild type (WT) controls. Methods WT and Z24 -/- mice received intramedullary fixed tibia fractures at ∼12 weeks of age. Mice were sacrificed throughout the time course of repair for the collection of organs that would provide information regarding the local (fracture callus, bone marrow, inguinal lymph nodes) versus peripheral (peripheral blood, contralateral tibia, abdominal organs) tissue microenvironments. Analyses of these specimens include histomorphometry, μCT, mechanical strength testing, protein quantification, gene expression analysis, flow cytometry for cellular senescence, and immunophenotyping. Results Z24 -/- mice demonstrated a significantly delayed rate of healing compared to WT mice with consistently smaller fracture calli containing higher proportion of cartilage and less bone after injury. Cellular senescence and pro-inflammatory cytokines were elevated in the Z24 -/- mice before and after fracture. These mice further presented with a dysregulated immune system, exhibiting generally decreased lymphopoiesis and increased myelopoiesis locally in the bone marrow, with more naïve and less memory T cell but greater myeloid activation systemically in the peripheral blood. Surprisingly, the ipsilateral lymph nodes had increased T cell activation and other pro-inflammatory NK and myeloid cells, suggesting that elevated myeloid abundance and activation contributes to an injury-specific hyperactivation of T cells. Conclusion Taken together, these data establish the Z24 -/- progeria mouse as a model of delayed fracture healing that exhibits decreased bone in the fracture callus, with weaker overall bone quality, immune dysregulation, and increased cellular senescence. Based on this mechanism for delayed healing, we propose this Z24 -/- progeria mouse model could be useful in testing novel therapeutics that could address delayed healing. The Translational Potential of this Article This study employs a novel animal model for delayed fracture healing that researchers can use to screen fracture healing therapeutics to address the globally prevalent issue of aberrant fracture healing.
Collapse
|
13
|
Nitschke BM, Beltran FO, Hahn MS, Grunlan MA. Trends in bioactivity: inducing and detecting mineralization of regenerative polymeric scaffolds. J Mater Chem B 2024; 12:2720-2736. [PMID: 38410921 PMCID: PMC10935659 DOI: 10.1039/d3tb02674d] [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: 11/10/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
Due to limitations of biological and alloplastic grafts, regenerative engineering has emerged as a promising alternative to treat bone defects. Bioactive polymeric scaffolds are an integral part of such an approach. Bioactivity importantly induces hydroxyapatite mineralization that promotes osteoinductivity and osseointegration with surrounding bone tissue. Strategies to confer bioactivity to polymeric scaffolds utilize bioceramic fillers, coatings and surface treatments, and additives. These approaches can also favorably impact mechanical and degradation properties. A variety of fabrication methods are utilized to prepare scaffolds with requisite morphological features. The bioactivity of scaffolds may be evaluated with a broad set of techniques, including in vitro (acellular and cellular) and in vivo methods. Herein, we highlight contemporary and emerging approaches to prepare and assess scaffold bioactivity, as well as existing challenges.
Collapse
Affiliation(s)
- Brandon M Nitschke
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA.
| | - Felipe O Beltran
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA.
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
14
|
Sirisereephap K, Tamura H, Lim JH, Surboyo MDC, Isono T, Hiyoshi T, Rosenkranz AL, Sato-Yamada Y, Domon H, Ikeda A, Hirose T, Sunazuka T, Yoshiba N, Okada H, Terao Y, Maeda T, Tabeta K, Chavakis T, Hajishengallis G, Maekawa T. A novel macrolide-Del-1 axis to regenerate bone in old age. iScience 2024; 27:108798. [PMID: 38261928 PMCID: PMC10797555 DOI: 10.1016/j.isci.2024.108798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/24/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
Aging is associated with increased susceptibility to chronic inflammatory bone loss disorders, such as periodontitis, in large part due to the impaired regenerative potential of aging tissues. DEL-1 exerts osteogenic activity and promotes bone regeneration. However, DEL-1 expression declines with age. Here we show that systemically administered macrolide antibiotics and a non-antibiotic erythromycin derivative, EM-523, restore DEL-1 expression in 18-month-old ("aged") mice while promoting regeneration of bone lost due to naturally occurring age-related periodontitis. These compounds failed to induce bone regeneration in age-matched DEL-1-deficient mice. Consequently, these drugs promoted DEL-1-dependent functions, including alkaline phosphatase activity and osteogenic gene expression in the periodontal tissue while inhibiting osteoclastogenesis, leading to net bone growth. Macrolide-treated aged mice exhibited increased skeletal bone mass, suggesting that this treatment may be pertinent to systemic bone loss disorders. In conclusion, we identified a macrolide-DEL-1 axis that can regenerate bone lost due to aging-related disease.
Collapse
Affiliation(s)
- Kridtapat Sirisereephap
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Hikaru Tamura
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meircurius Dwi Condro Surboyo
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Faculty of Dentistry, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Toshihito Isono
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Takumi Hiyoshi
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Andrea L. Rosenkranz
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Yurie Sato-Yamada
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Akari Ikeda
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoyasu Hirose
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Toshiaki Sunazuka
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Nagako Yoshiba
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Hiroyuki Okada
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Koichi Tabeta
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tomoki Maekawa
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| |
Collapse
|
15
|
Nelson AL, Fontana G, Chubb L, Choe J, Williams K, Regan D, Huard J, Murphy W, Ehrhart N, Bahney C. Mineral coated microparticles doped with fluoride and complexed with mRNA prolong transfection in fracture healing. Front Bioeng Biotechnol 2024; 11:1295313. [PMID: 38264578 PMCID: PMC10803474 DOI: 10.3389/fbioe.2023.1295313] [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: 09/16/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction: Impaired fracture healing, specifically non-union, has been found to occur up to 14% in tibial shaft fractures. The current standard of care to treat non-union often requires additional surgeries which can result in long recovery times. Injectable-based therapies to accelerate fracture healing have the potential to mitigate the need for additional surgeries. Gene therapies have recently undergone significant advancements due to developments in nanotechnology, which improve mRNA stability while reducing immunogenicity. Methods: In this study, we tested the efficacy of mineral coated microparticles (MCM) and fluoride-doped MCM (FMCM) to effectively deliver firefly luciferase (FLuc) mRNA lipoplexes (LPX) to the fracture site. Here, adult mice underwent a tibia fracture and stabilization method and all treatments were locally injected into the fracture. Level of osteogenesis and amount of bone formation were assessed using gene expression and histomorphometry respectively. Localized and systemic inflammation were measured through gene expression, histopathology scoring and measuring C-reactive protein (CRP) in the serum. Lastly, daily IVIS images were taken to track and measure transfection over time. Results: MCM-LPX-FLuc and FMCM-LPX-FLuc were not found to cause any cytotoxic effects when tested in vitro. When measuring the osteogenic potential of each mineral composition, FMCM-LPX-FLuc trended higher in osteogenic markers through qRT-PCR than the other groups tested in a murine fracture and stabilization model. Despite FMCM-LPX-FLuc showing slightly elevated il-1β and il-4 levels in the fracture callus, inflammation scoring of the fracture callus did not result in any differences. Additionally, an acute systemic inflammatory response was not observed in any of the samples tested. The concentration of MCM-LPX-FLuc and FMCM-LPX-FLuc that was used in the murine fracture model did not stimulate bone when analyzed through stereological principles. Transfection efficacy and kinetics of delivery platforms revealed that FMCM-LPX-FLuc prolongs the luciferase signal both in vitro and in vivo. Discussion: These data together reveal that FMCM-LPX-FLuc could serve as a promising mRNA delivery platform for fracture healing applications.
Collapse
Affiliation(s)
- Anna Laura Nelson
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Gianluca Fontana
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, United States
| | - Laura Chubb
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Josh Choe
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, United States
| | - Katherine Williams
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Colorado State University, Fort Collins, CO, United States
| | - Dan Regan
- Department of Microbiology, Colorado State University, Fort Collins, CO, United States
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - William Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Nicole Ehrhart
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Chelsea Bahney
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
- Orthopaedic Trauma Institute, University of California, San Francisco, CA, United States
| |
Collapse
|
16
|
da Rocha LR, Dias RB, Fernandes MBC, Prinz R, Eirado TP, Costa IDS, Monteiro MJ, da Silva CER, Dos Santos CT, Fogagnolo F. A new option for bone regeneration: a rapid methodology for cellularization of allograft with human bone marrow stromal cells with in vivo bone-forming potential. Injury 2023; 54 Suppl 6:110777. [PMID: 38143129 DOI: 10.1016/j.injury.2023.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 12/26/2023]
Abstract
The treatment of severe musculoskeletal injuries, such as loss of bone tissue and consolidation disorders, requires bone transplantation, and the success of this bone reconstruction depends on the grafts transplant's osteogenic, osteoconductive, and osteoinductive properties. Although the gold standard is autograft, it is limited by availability, morbidity, and infection risk. Despite their low capacity for osteoinduction and osteogenesis, decellularized bone allografts have been used in the search for alternative therapeutic strategies to improve bone regeneration. Considering that bone marrow stromal cells (BMSCs) are responsible for the maintenance of bone turnover throughout life, we believe that associating BMSCs with allograft could produce a material that is biologically similar to autologous bone graft. For this reason, this study evaluated the osteogenic potential of bone allograft cellularized with BMSCs. First, BMSC was characterized and allograft decellularization was confirmed by histology, scanning electron microscopy, and DNA quantification. Subsequently, the BMSCs and allografts were associated and evaluated for adhesion, proliferation, and in vitro and in vivo osteogenic potential. We demonstrated that, after 2 hours, BMSCs had already adhered to the surface of allografts and remained viable for 14 days. In vitro osteogenic assays indicated increased osteogenic potential of allografts compared with beta-tricalcium phosphate (β-TCP). In vivo transplantation assays in immunodeficient mice confirmed the allograft's potential to induce bone formation, with significantly better results than β-TCP. Finally, our results indicate that allograft can provide structural support for BMSC adhesion, offering a favorable microenvironment for cell survival and differentiation and inducing new bone formation. Taken together, our data indicate that this rapid methodology for cellularization of allograft with BMSCs might be a new therapeutic alternative in regenerative medicine and bone bioengineering.
Collapse
Affiliation(s)
- Leonardo Rosa da Rocha
- Teaching and Research Division, Instituto Nacional de Traumatologia e Ortopedia Jamil Haddad (INTO), Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil.
| | - Rhayra Braga Dias
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | | | - Rafael Prinz
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | - Thiago Penna Eirado
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | - Isabela de Souza Costa
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | - Mauricio J Monteiro
- Materials Division, Instituto Nacional de Tecnologia (INT), Av. Venezuela 82, Rio de Janeiro, RJ 20081-312, Brazil.
| | | | | | - Fabricio Fogagnolo
- Department of Orthopaedics and Anaesthesiology, Ribeirão Preto Medical School, Universidade de São Paulo (USP), Av. Bandeirantes, 3900, São Paulo, SP 14049900, Brazil
| |
Collapse
|
17
|
Qiao S, Zhang X, Chen Z, Zhao Y, Tzeng CM. Alloferon-1 ameliorates estrogen deficiency-induced osteoporosis through dampening the NLRP3/caspase-1/IL-1β/IL-18 signaling pathway. Int Immunopharmacol 2023; 124:110954. [PMID: 37742365 DOI: 10.1016/j.intimp.2023.110954] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/29/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023]
Abstract
Alloferon-1 is an insect polypeptide that has anti-inflammatory, antitumor and antiviral activity. This study aimed to determine the effects of alloferon-1 on estrogen deficiency-induced osteoporosis and explore the associated mechanism using a murine model of ovariectomy (OVX)-induced osteoporosis. Results showed that alloferon-1 prevented ovariectomy‑induced body weight gain, bone loss and bone mineral content reduction, affected biochemical markers of bone turnover, and restored the microstructure of bone trabeculae. Moreover, alloferon-1 suppressed the expression of the ovariectomy‑mediated inflammatory cytokines in the vertebrae bone tissues, including nucleotide-binding oligomerization domain-like-receptor family pyrin domain-containing 3 (NLRP3), cysteinyl aspartate specific proteinase-1 (Caspase-1), interleukin 1β (IL-1β) and interleukin 18 (IL-18) which were determined by immunofluorescence staining and western blot. Overall, the present study provides evidence for the effectiveness of alloferon-1 against estrogen deficiency-induced osteoporosis, suggesting an alternative drug or an auxiliary modulator for the treatment of postmenopausal osteoporosis (PMOP).
Collapse
Affiliation(s)
- Shuai Qiao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, Jiangsu, China
| | - Xiangrui Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, Jiangsu, China
| | - Ziyi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, Jiangsu, China
| | - Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, Jiangsu, China.
| | - Chi-Meng Tzeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, Fujian, China; Translational Medicine Research Center-Key Laboratory for Cancer T-Cell Theragnostic and Clinical Translation, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, Fujian, China; Xiamen Chang Gung Hospital Medical Research Center, Xiamen 361005, Fujian, China.
| |
Collapse
|
18
|
Davies BK, Hibbert AP, Roberts SJ, Roberts HC, Tickner JC, Holdsworth G, Arnett TR, Orriss IR. A Machine Learning-Based Image Segmentation Method to Quantify In Vitro Osteoclast Culture Endpoints. Calcif Tissue Int 2023; 113:437-448. [PMID: 37566229 PMCID: PMC10516805 DOI: 10.1007/s00223-023-01121-z] [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: 06/06/2023] [Accepted: 07/29/2023] [Indexed: 08/12/2023]
Abstract
Quantification of in vitro osteoclast cultures (e.g. cell number) often relies on manual counting methods. These approaches are labour intensive, time consuming and result in substantial inter- and intra-user variability. This study aimed to develop and validate an automated workflow to robustly quantify in vitro osteoclast cultures. Using ilastik, a machine learning-based image analysis software, images of tartrate resistant acid phosphatase-stained mouse osteoclasts cultured on dentine discs were used to train the ilastik-based algorithm. Assessment of algorithm training showed that osteoclast numbers strongly correlated between manual- and automatically quantified values (r = 0.87). Osteoclasts were consistently faithfully segmented by the model when visually compared to the original reflective light images. The ability of this method to detect changes in osteoclast number in response to different treatments was validated using zoledronate, ticagrelor, and co-culture with MCF7 breast cancer cells. Manual and automated counting methods detected a 70% reduction (p < 0.05) in osteoclast number, when cultured with 10 nM zoledronate and a dose-dependent decrease with 1-10 μM ticagrelor (p < 0.05). Co-culture with MCF7 cells increased osteoclast number by ≥ 50% irrespective of quantification method. Overall, an automated image segmentation and analysis workflow, which consistently and sensitively identified in vitro osteoclasts, was developed. Advantages of this workflow are (1) significantly reduction in user variability of endpoint measurements (93%) and analysis time (80%); (2) detection of osteoclasts cultured on different substrates from different species; and (3) easy to use and freely available to use along with tutorial resources.
Collapse
Affiliation(s)
- Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Andrew P Hibbert
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Scott J Roberts
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Helen C Roberts
- Department of Natural Sciences, Middlesex University, London, UK
| | - Jennifer C Tickner
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | | | - Timothy R Arnett
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
19
|
de Paula Colares Neto G, Pereira RMR, Alvarenga JC, Takayama L, de Assis Funari MF, Martin RM. Evaluation of the trabecular bone score in 35 children and adults with X-linked hypophosphatemic rickets. J Bone Miner Metab 2023; 41:666-672. [PMID: 37418074 DOI: 10.1007/s00774-023-01442-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 05/24/2023] [Indexed: 07/08/2023]
Abstract
INTRODUCTION The aim of this study is to evaluate and compare the trabecular bone scores (TBSs) of 11 children and 24 adults with X-linked hypophosphatemic rickets (XLH) and non-XLH subjects from a tertiary center. MATERIALS AND METHODS The areal bone mineral density at the lumbar spine (LS-aBMD) and LS-aBMD Z score were analyzed by dual-energy X-ray absorptiometry. The bone mineral apparent density (BMAD) and LS-aBMD Z score adjusted for height Z score (LS-aBMD-HAZ) were calculated. The TBS was determined using TBS iNsight software based on DXA images from the Hologic QDR 4500 device. RESULTS The XLH patients exhibited a higher mean LS-aBMD Z score, BMAD, and TBS than the non-XLH subjects (p < 0.01). LS-aBMD-HAZ and BMAD were greater in the XLH children than those in their corresponding non-XLH subjects (p < 0.01 and p = 0.02), and the XLH children trended toward a greater TBS (p = 0.06). The XLH adults had a higher LS-aBMD Z score, BMAD, and TBS than the non-XLH subjects (p < 0.01). When stratified by metabolic status according to the serum values of bone formation markers, compensated adult patients had a higher LS-aBMD Z score, BMAD, and TBS than non-XLH subjects (p < 0.01). Noncompensated patients had higher LS-aBMD Z scores and BMAD results than non-XLH subjects. However, TBS values did not differ statistically significantly between those groups (p = 0.45). CONCLUSION The higher LS-aBMD Z score, BMAD, and TBS result in the XLH patients compared to non-XLH subjects indicates an increased amount of trabecular bone within the lumbar spine, regardless of extraskeletal calcifications.
Collapse
Affiliation(s)
- Guido de Paula Colares Neto
- Osteometabolic Disorders Unit, Endocrinology Division, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 155-PAMB, 8° Andar, Bloco 3, São Paulo, SP, CEP: 05403-900, Brazil.
- Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 155-PAMB, 2° Andar, Bloco 6, São Paulo, SP, CEP: 05403-900, Brazil.
| | - Rosa Maria Rodrigues Pereira
- Bone Metabolism Laboratory (LIM/17), Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3º Andar, Sala 3193, São Paulo, SP, CEP: 01246-903, Brazil
| | - Jackeline Couto Alvarenga
- Bone Metabolism Laboratory (LIM/17), Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3º Andar, Sala 3193, São Paulo, SP, CEP: 01246-903, Brazil
| | - Liliam Takayama
- Bone Metabolism Laboratory (LIM/17), Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3º Andar, Sala 3193, São Paulo, SP, CEP: 01246-903, Brazil
| | - Mariana Ferreira de Assis Funari
- Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 155-PAMB, 2° Andar, Bloco 6, São Paulo, SP, CEP: 05403-900, Brazil
| | - Regina Matsunaga Martin
- Osteometabolic Disorders Unit, Endocrinology Division, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 155-PAMB, 8° Andar, Bloco 3, São Paulo, SP, CEP: 05403-900, Brazil
- Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 155-PAMB, 2° Andar, Bloco 6, São Paulo, SP, CEP: 05403-900, Brazil
| |
Collapse
|
20
|
Chandra Sekaran SP, Thotakura B, Jyothi AK, Manickam S, Chanemougavally J, Prabhu K, Gopalan DH. Effect of chlorpyrifos and its metabolites on skeletal system development of chick embryo. Birth Defects Res 2023; 115:1063-1078. [PMID: 37122261 DOI: 10.1002/bdr2.2170] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 05/02/2023]
Abstract
RESEARCH FOCUS Chlorpyrifos is an organophosphate insecticide used primarily to control pests on a variety of food and feed crops. Humans are directly or indirectly exposed to this pesticide through food, air, and occupation. The ill effects of chlorpyrifos on various organ systems of human has been widely documented, but less is known about its influence on human bones. AIM To analyze the effect of chlorpyrifos and its metabolites 3,5,6-trichloro-2-pyridinol (TCPy) on the skeletal system of the chick embryo. MATERIALS AND METHODS Fertilized chick eggs were exposed to different concentrations of chlorpyrifos and its metabolite 3,5,6-TCPy on 1.5 days of incubation. The proximal phalanx of 18-day-old embryos was analyzed for defects in growth and ossification through histopathology, immunohistochemistry, angiogenesis assay, and gene expression study. RESULTS Dose-dependent variations in developing bone of chick embryo were observed. Histochemical and histomorphometry studies of proximal phalanx showed increased in the growth plate length (F(9, 59) = 228.9509, p = .00001) with a reduction in the total length of the phalanx (F(9, 59) = 109.9905, p = .00001), decreased mineralization (F(9, 59) = 224.6872, p = .00001), decreased blood islands in the bone marrow (F(9, 59) = 7.7083, p = .0001) of chlorpyrifos, and 3,5,6-TCPy-exposed group. Significant downregulations in the expression patterns of the transcription factors, such as SOX9, RUNX2, and ALP, were also observed. CONCLUSION Chlorpyrifos and its metabolite 3,5,6-TCPy exposure alters the chondrogenesis in the growth plate cartilage of long bone in chick embryo. The pesticide and its metabolite also interfere in ossification.
Collapse
Affiliation(s)
| | - Balaji Thotakura
- Department of Anatomy, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | - Ashok Kumar Jyothi
- Apollo Institute of Medical Sciences & Research, Chittoor, Andhrapradesh, India
| | - Subramanian Manickam
- Department of Anatomy, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | - Jayaprakash Chanemougavally
- Department of Anatomy, A.C.S Medical College and Hospital, Dr. MGR Educational and Research Institute Velappanchavadi, Chennai, Tamil Nadu, India
| | - Kaliaperumal Prabhu
- Department of Anatomy, Sree Balaji Medical College and Hospital, Chennai, Tamil Nadu, India
| | | |
Collapse
|
21
|
Storlino G, Dicarlo M, Zerlotin R, Pignataro P, Sanesi L, Suriano C, Oranger A, Mori G, Passeri G, Colucci S, Grano M, Colaianni G. Irisin Protects against Loss of Trabecular Bone Mass and Strength in Adult Ovariectomized Mice by Stimulating Osteoblast Activity. Int J Mol Sci 2023; 24:9896. [PMID: 37373043 DOI: 10.3390/ijms24129896] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Irisin is a peptide secreted by skeletal muscle that plays a major role in bone metabolism. Experiments in mouse models have shown that administration of recombinant irisin prevents disuse-induced bone loss. In this study, we aimed to evaluate the effects of irisin treatment for the prevention of bone loss in the ovariectomized (Ovx) mouse, the animal model commonly used to investigate osteoporosis caused by estrogen deficiency. Micro-Ct analysis conducted on Sham mice (Sham-veh) and Ovx mice treated with vehicle (Ovx-veh) or recombinant irisin (Ovx-irisn) showed bone volume fraction (BV/TV) decreases in femurs (Ovx-veh 1.39± 0.71 vs. Sham-veh 2.84 ± 1.23; p = 0.02) and tibia at both proximal condyles (Ovx-veh 1.97 ± 0.68 vs. Sham-veh 3.48 ± 1.26; p = 0.03) and the subchondral plate (Ovx-veh 6.33 ± 0.36 vs. Sham-veh 8.18 ± 0.41; p = 0.01), which were prevented by treatment with a weekly dose of irisin for 4 weeks. Moreover, histological analysis of trabecular bone showed that irisin increased the number of active osteoblasts per bone perimeter (Ovx-irisin 32.3 ± 3.9 vs. Ovx-veh 23.5 ± 3.6; p = 0.01), while decreasing osteoclasts (Ovx-irisin 7.6 ± 2.4 vs. Ovx-veh 12.9 ± 3.04; p = 0.05). The possible mechanism by which irisin enhances osteoblast activity in Ovx mice is upregulation of the transcription factor Atf4, one of the key markers of osteoblast differentiation, and osteoprotegerin, thereby inhibiting osteoclast formation.
Collapse
Affiliation(s)
- Giuseppina Storlino
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
| | - Manuela Dicarlo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Roberta Zerlotin
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Patrizia Pignataro
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy
| | - Lorenzo Sanesi
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy
| | - Clelia Suriano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Angela Oranger
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
| | - Giovanni Passeri
- Unit of Clinica e Terapia Medica, Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| | - Silvia Colucci
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| |
Collapse
|
22
|
Rivera KO, Cuylear DL, Duke VR, O’Hara KM, Zhong JX, Elghazali NA, Finbloom JA, Kharbikar BN, Kryger AN, Miclau T, Marcucio RS, Bahney CS, Desai TA. Encapsulation of β-NGF in injectable microrods for localized delivery accelerates endochondral fracture repair. Front Bioeng Biotechnol 2023; 11:1190371. [PMID: 37284244 PMCID: PMC10241161 DOI: 10.3389/fbioe.2023.1190371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/02/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction: Currently, there are no non-surgical FDA-approved biological approaches to accelerate fracture repair. Injectable therapies designed to stimulate bone healing represent an exciting alternative to surgically implanted biologics, however, the translation of effective osteoinductive therapies remains challenging due to the need for safe and effective drug delivery. Hydrogel-based microparticle platforms may be a clinically relevant solution to create controlled and localized drug delivery to treat bone fractures. Here, we describe poly (ethylene glycol) dimethacrylate (PEGDMA)-based microparticles, in the shape of microrods, loaded with beta nerve growth factor (β-NGF) for the purpose of promoting fracture repair. Methods: Herein, PEGDMA microrods were fabricated through photolithography. PEGDMA microrods were loaded with β-NGF and in vitro release was examined. Subsequently, bioactivity assays were evaluated in vitro using the TF-1 tyrosine receptor kinase A (Trk-A) expressing cell line. Finally, in vivo studies using our well-established murine tibia fracture model were performed and a single injection of the β-NGF loaded PEGDMA microrods, non-loaded PEGDMA microrods, or soluble β-NGF was administered to assess the extent of fracture healing using Micro-computed tomography (µCT) and histomorphometry. Results: In vitro release studies showed there is significant retention of protein within the polymer matrix over 168 hours through physiochemical interactions. Bioactivity of protein post-loading was confirmed with the TF-1 cell line. In vivo studies using our murine tibia fracture model show that PEGDMA microrods injected at the site of fracture remained adjacent to the callus for over 7 days. Importantly, a single injection of β-NGF loaded PEGDMA microrods resulted in improved fracture healing as indicated by a significant increase in the percent bone in the fracture callus, trabecular connective density, and bone mineral density relative to soluble β-NGF control indicating improved drug retention within the tissue. The concomitant decrease in cartilage fraction supports our prior work showing that β-NGF promotes endochondral conversion of cartilage to bone to accelerate healing. Discussion: We demonstrate a novel and translational method wherein β-NGF can be encapsulated within PEGDMA microrods for local delivery and that β-NGF bioactivity is maintained resulting in improved bone fracture repair.
Collapse
Affiliation(s)
- Kevin O. Rivera
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Darnell L. Cuylear
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Victoria R. Duke
- Center for Regenerative and Personalized Medicine, The Steadman Philippon Research Institute (SPRI), Vail, CO, United States
| | - Kelsey M. O’Hara
- Center for Regenerative and Personalized Medicine, The Steadman Philippon Research Institute (SPRI), Vail, CO, United States
| | - Justin X. Zhong
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
- UC Berkeley—UCSF Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Nafisa A. Elghazali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
- UC Berkeley—UCSF Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Joel A. Finbloom
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Bhushan N. Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Alex N. Kryger
- School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Chelsea S. Bahney
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Center for Regenerative and Personalized Medicine, The Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- UC Berkeley—UCSF Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Tejal A. Desai
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering, University of California, Berkeley (UC Berkeley), Berkeley, CA, United States
- School of Engineering, Brown University, Providence, RI, United States
| |
Collapse
|
23
|
de Souza Balbinot G, Leitune VCB, da Cunha Bahlis EA, Ponzoni D, Visioli F, Collares FM. Niobium-containing bioactive glasses modulate alkaline phosphatase activity during bone repair. J Biomed Mater Res B Appl Biomater 2023; 111:1224-1231. [PMID: 36773168 DOI: 10.1002/jbm.b.35227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/12/2023]
Abstract
This study aimed to evaluate the pre-clinical behavior of niobium-containing bioactive glasses (BAGNb) by their ability to promote bone repair and regulate alkaline phosphatase (ALP) levels in an animal model. BAGNbs were produced as powders and as scaffolds and surgically implanted in the femur of male rats (Wistar lineage n = 10). Glasses without Nb (BAG) were produced and implanted as well. The Autogenous Bone (AB) was used as a control. After 15, 30, and 60 days of surgical implantation, blood serum samples were collected to quantify ALP activity, and femurs were removed to assess bone repair. Bone samples were histologically processed and stained with H&E to quantify the % new bone into defects. No postoperative complications were identified. Early-stage repair (15 days) resulted in increased ALP activity for all groups, with increased values for powdered BAGNb. The maturation of the new bone led to a reduction in serum ALP levels. Histological sections showed the formation of immature bone tissue and vascularization with the progression of bone deposition to mature and functional tissue over time. BAG powder showed less new bone formation in 15 days, while the analysis at 30 and 60 days showed no difference between groups (p > .05). Niobium-containing bioactive glasses safely and successfully induced bone repair in vivo. The modulation of ALP activity may be a pathway to describe the ability of niobium-containing materials to contribute to new bone formation.
Collapse
Affiliation(s)
- Gabriela de Souza Balbinot
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Deise Ponzoni
- Oral and Maxillofacial Surgery Unit, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Visioli
- Department of Oral Pathology, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabrício Mezzomo Collares
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
24
|
Landscape of Well-Coordinated Fracture Healing in a Mouse Model Using Molecular and Cellular Analysis. Int J Mol Sci 2023; 24:ijms24043569. [PMID: 36834981 PMCID: PMC9964763 DOI: 10.3390/ijms24043569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The success of fracture healing relies on overlapping but coordinated cellular and molecular events. Characterizing an outline of differential gene regulation throughout successful healing is essential for identifying crucial phase-specific markers and may serve as the basis for engineering these in challenging healing situations. This study analyzed the healing progression of a standard closed femoral fracture model in C57BL/6N (age = 8 weeks) wild-type male mice. The fracture callus was assessed across various days post fracture (D = days 0, 3, 7, 10, 14, 21, and 28) by microarray, with D0 serving as a control. Histological analyses were carried out on samples from D7 until D28 to support the molecular findings. Microarray analysis revealed a differential regulation of immune response, angiogenesis, ossification, extracellular matrix regulation, mitochondrial and ribosomal genes during healing. In-depth analysis showed differential regulation of mitochondrial and ribosomal genes during the initial phase of healing. Furthermore, the differential gene expression showed an essential role of Serpin Family F Member 1 over the well-known Vascular Endothelial Growth Factor in angiogenesis, especially during the inflammatory phase. The significant upregulation of matrix metalloproteinase 13 and bone sialoprotein from D3 until D21 asserts their importance in bone mineralization. The study also shows type I collagen around osteocytes located in the ossified region at the periosteal surface during the first week of healing. Histological analysis of matrix extracellular phosphoglycoprotein and extracellular signal-regulated kinase stressed their roles in bone homeostasis and the physiological bone-healing process. This study reveals previously unknown and novel candidates, that could serve as a target for specific time points in healing and to remedy cases of impaired healing.
Collapse
|
25
|
Thieu MKL, Stoetzel S, Rahmati M, El Khassawna T, Verket A, Sanz-Esporrin J, Sanz M, Ellingsen JE, Haugen HJ. Immunohistochemical comparison of lateral bone augmentation using a synthetic TiO 2 block or a xenogeneic graft in chronic alveolar defects. Clin Implant Dent Relat Res 2023; 25:57-67. [PMID: 36222116 PMCID: PMC10092822 DOI: 10.1111/cid.13143] [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: 07/14/2022] [Revised: 08/28/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To evaluate osteogenic markers and alveolar ridge profile changes in guided bone regeneration (GBR) of chronic noncontained bone defects using a nonresorbable TiO2 block. MATERIALS AND METHODS Three buccal bone defects were created in each hemimandible of eight beagle dogs and allowed to heal for 8 weeks before GBR. Treatment was assigned by block randomization: TiO2 block: TiO2 -scaffold and a collagen membrane, DBBM particulates: Deproteinized bovine bone mineral (DBBM) and a collagen membrane, Empty control: Only collagen membrane. Bone regeneration was assessed on two different healing timepoints: early (4 weeks) and late healing (12 weeks) using several immunohistochemistry markers including alpha-smooth muscle actin (α-SMA), osteopontin, osteocalcin, tartrate-resistant acid phosphatase, and collagen type I. Histomorphometry was performed on Movat Pentachrome-stained and Von Kossa/Van Gieson-stained sections. Stereolithographic (STL) models were used to compare alveolar profile changes. RESULTS The percentage of α-SMA and osteopontin increased in TiO2 group after 12 weeks of healing at the bone-scaffold interface, while collagen type I increased in the empty control group. In the defect area, α-SMA decreased in the empty control group, while collagen type I increased in the DBBM group. All groups maintained alveolar profile from 4 to 12 weeks, but TiO2 group demonstrated the widest soft tissue contour profile. CONCLUSIONS The present findings suggested contact osteogenesis when GBR is performed with a TiO2 block or DBBM particulates. The increase in osteopontin indicated a potential for bone formation beyond 12 weeks. The alveolar profile data indicated a sustained lateral increase in lateral bone augmentation using a TiO2 block and a collagen membrane, as compared with DBBM and a collagen membrane or a collagen membrane alone.
Collapse
Affiliation(s)
- Minh Khai Le Thieu
- Department of Periodontology, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway.,Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Sabine Stoetzel
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Maryam Rahmati
- Department of Periodontology, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Thaqif El Khassawna
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Anders Verket
- Department of Experimental Trauma Surgery, Faculty of Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | | | - Mariano Sanz
- Periodontology, University Complutense of Madrid, Madrid, Spain
| | - Jan Eirik Ellingsen
- Department of Prosthetics and Oral Function, University of Oslo, Oslo, Norway
| | - Håvard Jostein Haugen
- Department of Periodontology, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
26
|
Brent MB, Emmanuel T. Contemporary Advances in Computer-Assisted Bone Histomorphometry and Identification of Bone Cells in Culture. Calcif Tissue Int 2023; 112:1-12. [PMID: 36309622 DOI: 10.1007/s00223-022-01035-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/13/2022] [Indexed: 01/07/2023]
Abstract
Static and dynamic bone histomorphometry and identification of bone cells in culture are labor-intensive and highly repetitive tasks. Several computer-assisted methods have been proposed to ease these tasks and to take advantage of the increased computational power available today. The present review aimed to provide an overview of contemporary methods utilizing specialized computer software to perform bone histomorphometry or identification of bone cells in culture. In addition, a brief historical perspective on bone histomorphometry is included. We identified ten publications using five different computer-assisted approaches (1) ImageJ and BoneJ; (2) Histomorph: OsteoidHisto, CalceinHisto, and TrapHisto; (3) Fiji/ImageJ2 and Trainable Weka Segmentation (TWS); (4) Visiopharm and artificial intelligence (AI); and (5) Osteoclast identification using deep learning with Single Shot Detection (SSD) architecture, Darknet and You Only Look Once (YOLO), or watershed algorithm (OC_Finder). The review also highlighted a substantial need for more validation studies that evaluate the accuracy of the new computational methods to the manual and conventional analyses of histological bone specimens and cells in culture using microscopy. However, a substantial evolution has occurred during the last decade to identify and separate bone cells and structures of interest. Most early studies have used simple image segmentation to separate structures of interest, whereas the most recent studies have utilized AI and deep learning. AI has been proposed to substantially decrease the amount of time needed for analyses and enable unbiased assessments. Despite the clear advantages of highly sophisticated computational methods, the limited nature of existing validation studies, particularly those that assess the accuracy of the third-generation methods compared to the second-generation methods, appears to be an important reason that these techniques have failed to gain wide acceptance.
Collapse
Affiliation(s)
- Mikkel Bo Brent
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 3, 8000, Aarhus, Denmark.
| | - Thomas Emmanuel
- Department of Dermatology, Aarhus University Hospital, 8200, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University Hospital, 8200, Aarhus, Denmark
| |
Collapse
|
27
|
Alcorta-Sevillano N, Infante A, Macías I, Rodríguez CI. Murine Animal Models in Osteogenesis Imperfecta: The Quest for Improving the Quality of Life. Int J Mol Sci 2022; 24:ijms24010184. [PMID: 36613624 PMCID: PMC9820162 DOI: 10.3390/ijms24010184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
Osteogenesis imperfecta is a rare genetic disorder characterized by bone fragility, due to alterations in the type I collagen molecule. It is a very heterogeneous disease, both genetically and phenotypically, with a high variability of clinical phenotypes, ranging from mild to severe forms, the most extreme cases being perinatal lethal. There is no curative treatment for OI, and so great efforts are being made in order to develop effective therapies. In these attempts, the in vivo preclinical studies are of paramount importance; therefore, serious analysis is required to choose the right murine OI model able to emulate as closely as possible the disease of the target OI population. In this review, we summarize the features of OI murine models that have been used for preclinical studies until today, together with recently developed new murine models. The bone parameters that are usually evaluated in order to determine the relevance of new developing therapies are exposed, and finally, current and innovative therapeutic strategies attempts considered in murine OI models, along with their mechanism of action, are reviewed. This review aims to summarize the in vivo studies developed in murine models available in the field of OI to date, in order to help the scientific community choose the most accurate OI murine model when developing new therapeutic strategies capable of improving the quality of life.
Collapse
Affiliation(s)
- Natividad Alcorta-Sevillano
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
- Department of Cell Biology and Histology, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
| | - Iratxe Macías
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
| | - Clara I. Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
- Correspondence:
| |
Collapse
|
28
|
Oniszczuk A, Kaczmarek A, Kaczmarek M, Ciałowicz M, Arslan E, Silva AF, Clemente FM, Murawska-Ciałowicz E. Sclerostin as a biomarker of physical exercise in osteoporosis: A narrative review. Front Endocrinol (Lausanne) 2022; 13:954895. [PMID: 36545331 PMCID: PMC9760825 DOI: 10.3389/fendo.2022.954895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/14/2022] [Indexed: 12/07/2022] Open
Abstract
Osteoporosis, a disease of low bone mass, is characterized by reduced bone mineral density (BMD) through abnormalities in the microarchitecture of bone tissue. It affects both the social and economic areas, therefore it has been considered a lifestyle disease for many years. Bone tissue is a dynamic structure exhibiting sensitivity to various stimuli, including mechanical ones, which are a regulator of tissue sclerostin levels. Sclerostin is a protein involved in bone remodeling, showing an anti-anabolic effect on bone density. Moderate to vigorous physical activity inhibits secretion of this protein and promotes increased bone mineral density. Appropriate exercise has been shown to have an osteogenic effect. The effectiveness of osteogenic training depends on the type, intensity, regularity and frequency of exercise and the number of body parts involved. The greatest osteogenic activity is demonstrated by exercises affecting bone with high ground reaction forces (GRF) and high forces exerted by contracting muscles (JFR). The purpose of this study was to review the literature for the effects of various forms of exercise on sclerostin secretion.
Collapse
Affiliation(s)
- Anna Oniszczuk
- Department of Physiology and Biochemistry, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| | - Agnieszka Kaczmarek
- Department of Physiology and Biochemistry, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| | - Mateusz Kaczmarek
- Gynecology and Obstetrics Department, St. Hedwig’s of Silesia Hospital, Trzebnica, Poland
| | - Maria Ciałowicz
- Physiotherapy Faculty, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| | - Ersan Arslan
- Faculty of Sport Science, Gaziosmanpaşa University, Tokat, Turkey
| | - Ana Filipa Silva
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Rua Escola Industrial e Comercial de Nun’Álvares, Viana do Castelo, Portugal
- Research Center in Sports Performance, Recreation, Innovation and Technology (SPRINT), Melgaço, Portugal
- The Research Centre in Sports Sciences, Health Sciences and Human Development (CIDESD), Vila Real, Portugal
| | - Filipe Manuel Clemente
- Faculty of Sport Science, Gaziosmanpaşa University, Tokat, Turkey
- Research Center in Sports Performance, Recreation, Innovation and Technology (SPRINT), Melgaço, Portugal
| | - Eugenia Murawska-Ciałowicz
- Department of Physiology and Biochemistry, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| |
Collapse
|
29
|
Behera J, Ison J, Voor MJ, Tyagi N. Exercise-Linked Skeletal Irisin Ameliorates Diabetes-Associated Osteoporosis by Inhibiting the Oxidative Damage-Dependent miR-150-FNDC5/Pyroptosis Axis. Diabetes 2022; 71:2777-2792. [PMID: 35802043 PMCID: PMC9750954 DOI: 10.2337/db21-0573] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 06/22/2022] [Indexed: 01/25/2023]
Abstract
Recent evidence suggests that physical exercise (EX) promotes skeletal development. However, the impact of EX on the progression of bone loss and deterioration of mechanical strength in mice with type 2 diabetic mellitus (T2DM) remains unexplored. In the current study, we investigated the effect of EX on bone mass and mechanical quality using a diabetic mouse model. The T2DM mouse model was established with a high-fat diet with two streptozotocin injections (50 mg/kg/body wt) in C57BL/6 female mice. The diabetic mice underwent treadmill exercises (5 days/week at 7-11 m/min for 60 min/day) for 8 weeks. The data showed that diabetes upregulated miR-150 expression through oxidative stress and suppressed FNDC5/Irisin by binding to its 3'-untranslated region. The decreased level of irisin further triggers the pyroptosis response in diabetic bone tissue. EX or N-acetyl cysteine or anti-miRNA-150 transfection in T2DM mice restored FNDC5/Irisin expression and bone formation. Furthermore, EX or recombinant irisin administration prevented T2DM-Induced hyperglycemia and improved glucose intolerance in diabetic mice. Furthermore, osteoblastic knockdown of Nlrp3 silencing (si-Nlrp3) or pyroptosis inhibitor (Ac-YVADCMK [AYC]) treatment restores bone mineralization in diabetic mice. Micro-computed tomography scans and mechanical testing revealed that trabecular bone microarchitecture and bone mechanical properties were improved after EX in diabetic mice. Irisin, either induced by skeleton or daily EX or directly administered, prevents bone loss by mitigating inflammasome-associated pyroptosis signaling in diabetic mice. This study demonstrates that EX-induced skeletal irisin ameliorates diabetes-associated glucose intolerance and bone loss and possibly provides a mechanism of its effects on metabolic osteoporosis.
Collapse
Affiliation(s)
- Jyotirmaya Behera
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY
| | - Jessica Ison
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY
| | - Michael J. Voor
- Departments of Orthopaedic Surgery and Bioengineering, School of Medicine, University of Louisville, Louisville, KY
- Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY
| | - Neetu Tyagi
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY
| |
Collapse
|
30
|
Sun Y, Helmholz H, Willumeit-Römer R. Multicolor Histochemical Staining for Identification of Mineralized and Non-Mineralized Musculoskeletal Tissue: Immunohistochemical and Radiological Validation in Decalcified Bone Samples. Bioengineering (Basel) 2022; 9:bioengineering9100488. [PMID: 36290456 PMCID: PMC9598171 DOI: 10.3390/bioengineering9100488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
Histochemical staining of paraffin-embedded decalcified bone samples is commonly used in preclinical research of musculoskeletal diseases, enabling the visualization of multiple tissue components by the application of chromogens. The purpose of this study was to introduce a novel multicolor staining protocol involving optimized chemical reagents and procedure, allowing the identification of high-mineralized bone, low-mineralized fracture callus, cartilage and skeletal muscle fibers simultaneously. Fractured femur and healthy tail vertebra samples from adult male Sprague–Dawley rats were decalcified with EDTA and formic acid, respectively, followed by paraffin embedding, tissue sectioning and multicolor staining. Conventional Movat’s pentachrome and safranin O / fast green staining were conducted in parallel for comparison. Immunohistochemical staining of collagen type-X and micro-CT analysis were included to further validate the efficacy of the staining method. The multicolor staining allowed visualization of major musculoskeletal tissue components in both types of decalcified samples, providing quality outcomes with fewer chemical reagents and simplified procedures. Immunohistochemical staining demonstrated its capacity for identification of the endochondral ossification process during fracture healing. Micro-CT imaging validated the staining outcome for high-mineralized skeletal tissue. The application of the multicolor staining may facilitate future preclinical research involving decalcified paraffin-embedded samples.
Collapse
Affiliation(s)
- Yu Sun
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon, 21502 Geesthacht, Germany
- Department of Orthopaedics, First Hospital of China Medical University, Shenyang 110001, China
- Correspondence:
| | - Heike Helmholz
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon, 21502 Geesthacht, Germany
| | | |
Collapse
|
31
|
Licini C, Notarstefano V, Marchi S, Cerqueni G, Ciapetti G, Vitale‐Brovarone C, Giorgini E, Mattioli‐Belmonte M. Altered type I collagen networking in osteoporotic human femoral head revealed by histomorphometric and Fourier transform infrared imaging correlated analyses. Biofactors 2022; 48:1089-1110. [PMID: 35661288 PMCID: PMC9796100 DOI: 10.1002/biof.1870] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/30/2022] [Indexed: 12/30/2022]
Abstract
Bone homeostasis is the equilibrium between organic and inorganic components of the extracellular matrix (ECM) and cells. Alteration of this balance has consequences on bone mass and architecture, resulting in conditions such as osteoporosis (OP). Given ECM protein mutual regulation and their effects on bone structure and mineralization, further insight into their expression is crucial to understanding bone biology under normal and pathological conditions. This study focused on Type I Collagen, which is mainly responsible for structural properties and mineralization of bone, and selected proteins implicated in matrix composition, mineral deposition, and cell-matrix interaction such as Decorin, Osteocalcin, Osteopontin, Bone Sialoprotein 2, Osteonectin and Transforming Growth Factor beta. We developed a novel multidisciplinary approach in order to assess bone matrix in healthy and OP conditions more comprehensively by exploiting the Fourier Transform Infrared Imaging (FTIRI) technique combined with histomorphometry, Sirius Red staining, immunohistochemistry, and Western Blotting. This innovatory procedure allowed for the analysis of superimposed tissue sections and revealed that the alterations in OP bone tissue architecture were associated with warped Type I Collagen structure and deposition but not with changes in the total protein amount. The detected changes in the expression and/or cooperative or antagonist role of Decorin, Osteocalcin, Osteopontin, and Bone Sialoprotein-2 indicate the deep impact of these NCPs on collagen features of OP bone. Overall, our strategy may represent a starting point for designing targeted clinical strategies aimed at bone mass preservation and sustain the FTIRI translational capability as upcoming support for traditional diagnostic methods.
Collapse
Affiliation(s)
- Caterina Licini
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
- Department of Applied Science and TechnologyPolitecnico di TorinoTorinoItaly
| | - Valentina Notarstefano
- Department of Life and Environmental SciencesUniversità Politecnica delle MarcheAnconaItaly
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
| | - Giorgia Cerqueni
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
| | - Gabriela Ciapetti
- Laboratory of Nanobiotechnology (NaBi)IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | | | - Elisabetta Giorgini
- Department of Life and Environmental SciencesUniversità Politecnica delle MarcheAnconaItaly
| | - Monica Mattioli‐Belmonte
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
| |
Collapse
|
32
|
Harnett MM, Doonan J, Lumb FE, Crowe J, Damink RO, Buitrago G, Duncombe-Moore J, Wilkinson DI, Suckling CJ, Selman C, Harnett W. The parasitic worm product ES-62 protects the osteoimmunology axis in a mouse model of obesity-accelerated ageing. Front Immunol 2022; 13:953053. [PMID: 36105811 PMCID: PMC9465317 DOI: 10.3389/fimmu.2022.953053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite significant increases in human lifespan over the last century, adoption of high calorie diets (HCD) has driven global increases in type-2 diabetes, obesity and cardiovascular disease, disorders precluding corresponding improvements in healthspan. Reflecting that such conditions are associated with chronic systemic inflammation, evidence is emerging that infection with parasitic helminths might protect against obesity-accelerated ageing, by virtue of their evolution of survival-promoting anti-inflammatory molecules. Indeed, ES-62, an anti-inflammatory secreted product of the filarial nematode Acanthocheilonema viteae, improves the healthspan of both male and female C57BL/6J mice undergoing obesity-accelerated ageing and also extends median lifespan in male animals, by positively impacting on inflammatory, adipose metabolic and gut microbiome parameters of ageing. We therefore explored whether ES-62 affects the osteoimmunology axis that integrates environmental signals, such as diet and the gut microbiome to homeostatically regulate haematopoiesis and training of immune responses, which become dysregulated during (obesity-accelerated) ageing. Of note, we find sexual dimorphisms in the decline in bone health, and associated dysregulation of haematopoiesis and consequent peripheral immune responses, during obesity-accelerated ageing, highlighting the importance of developing sex-specific anti-ageing strategies. Related to this, ES-62 protects trabecular bone structure, maintaining bone marrow (BM) niches that counter the ageing-associated decline in haematopoietic stem cell (HSC) functionality highlighted by a bias towards myeloid lineages, in male but not female, HCD-fed mice. This is evidenced by the ability of ES-62 to suppress the adipocyte and megakaryocyte bias and correspondingly promote increases in B lymphocytes in the BM. Furthermore, the consequent prevention of ageing-associated myeloid/lymphoid skewing is associated with reduced accumulation of inflammatory CD11c+ macrophages and IL-1β in adipose tissue, disrupting the perpetuation of inflammation-driven dysregulation of haematopoiesis during obesity-accelerated ageing in male HCD-fed mice. Finally, we report the ability of small drug-like molecule analogues of ES-62 to mimic some of its key actions, particularly in strongly protecting trabecular bone structure, highlighting the translational potential of these studies.
Collapse
Affiliation(s)
- Margaret M. Harnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - James Doonan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Felicity E. Lumb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Jenny Crowe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Roel Olde Damink
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Geraldine Buitrago
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Josephine Duncombe-Moore
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Debbie I. Wilkinson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Colin J. Suckling
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
33
|
Rosch S, Kresoja KP, Besler C, Fengler K, Schöber AR, von Roeder M, Lücke C, Gutberlet M, Klingel K, Thiele H, Rommel KP, Lurz P. Characteristics of Heart Failure With Preserved Ejection Fraction Across the Range of Left Ventricular Ejection Fraction. Circulation 2022; 146:506-518. [PMID: 35862208 DOI: 10.1161/circulationaha.122.059280] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Recent trial data suggest that stratification of patients with heart failure with preserved ejection fraction (HFpEF) according to left ventricular ejection fraction (LVEF) provides a means for dissecting different treatment responses. However, the differential pathophysiologic considerations have rarely been described. METHODS This prospective, single-center study analyzed consecutive symptomatic patients with HFpEF diagnosed according to the 2016 European Society of Cardiology heart failure guidelines. Patients were grouped into LVEF 50% to 60% and LVEF >60% cohorts. All patients underwent cardiac magnetic resonance imaging. Transfemoral cardiac catheterization was performed to derive load-dependent and load-independent left ventricular (LV) properties on pressure-volume loop analyses. RESULTS Fifty-six patients with HFpEF were enrolled and divided into LVEF 50% to 60% (n=21) and LVEF >60% (n=35) cohorts. On cardiac magnetic resonance imaging, the LVEF >60% cohort showed lower LV end-diastolic volumes (P=0.019) and end-systolic volumes (P=0.001) than the LVEF 50% to 60% cohort; stroke volume (P=0.821) did not differ between the cohorts. Extracellular volume fraction was higher in the LVEF 50% to 60% cohort than in the LVEF >60% cohort (0.332 versus 0.309; P=0.018). Pressure-volume loop analyses demonstrated higher baseline LV contractility (end-systolic elastance, 1.85 vs 1.33 mm Hg/mL; P<0.001) and passive diastolic stiffness (β constant, 0.032 versus 0.018; P=0.004) in the LVEF >60% cohort. Ventriculo-arterial coupling (end-systolic elastance/arterial elastance) at rest was in the range of optimized stroke work in the LVEF >60% cohort but was impaired in the LVEF 50% to 60% cohort (1.01 versus 0.80; P=0.005). During handgrip exercise, patients with LVEF >60% had higher increases in end-systolic elastance (1.85 versus 0.82 mm Hg/mL; P=0.023), attenuated increases in indexed end-systolic volume (-1 versus 7 mL/m²; P<0.004), and more exaggerated increases in LV filling pressures (8 vs 5 mm Hg; P=0.023). LV stroke volume decreased in the LVEF >60% cohort (P=0.007) under exertion. CONCLUSIONS Patients with HFpEF in whom LVEF ranged from 50% to 60% demonstrated reduced contractility, impaired ventriculo-arterial coupling, and higher extracellular volume fraction. In contrast, patients with HFpEF and a LVEF >60% demonstrated a hypercontractile state with excessive LV afterload and diminished preload reserve. A LVEF-based stratification of patients with HFpEF identified distinct morphologic and pathophysiologic subphenotypes.
Collapse
Affiliation(s)
- Sebastian Rosch
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Karl-Patrik Kresoja
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Christian Besler
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Karl Fengler
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Anne Rebecca Schöber
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Maximilian von Roeder
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Christian Lücke
- Radiology (C.L., M.G.), Heart Center Leipzig at University of Leipzig, Germany
| | - Matthias Gutberlet
- Radiology (C.L., M.G.), Heart Center Leipzig at University of Leipzig, Germany
| | - Karin Klingel
- Department of Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Germany (K.K.)
| | - Holger Thiele
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Karl-Philipp Rommel
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| | - Philipp Lurz
- Departments of Cardiology (S.R., K.-P.K., C.B., K.F., A.R.S., M.v.R., H.T., K.-P.R., P.L.), Heart Center Leipzig at University of Leipzig, Germany
| |
Collapse
|
34
|
Mohammadpour A, Mahabady MK, Ranjbar R, Tabandeh MR, Jamshidian J. Effect of Mentha spicata L. Essential Oil Orally Exposure During Organogenesis in Wistar Rats and Development of Fetus Bone. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02626-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Mohd N, Razali M, Ghazali MJ, Abu Kasim NH. 3D-Printed Hydroxyapatite and Tricalcium Phosphates-Based Scaffolds for Alveolar Bone Regeneration in Animal Models: A Scoping Review. MATERIALS (BASEL, SWITZERLAND) 2022; 15:2621. [PMID: 35407950 PMCID: PMC9000240 DOI: 10.3390/ma15072621] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/18/2022] [Accepted: 03/29/2022] [Indexed: 02/07/2023]
Abstract
Three-dimensional-printed scaffolds have received greater attention as an attractive option compared to the conventional bone grafts for regeneration of alveolar bone defects. Hydroxyapatite and tricalcium phosphates have been used as biomaterials in the fabrication of 3D-printed scaffolds. This scoping review aimed to evaluate the potential of 3D-printed HA and calcium phosphates-based scaffolds on alveolar bone regeneration in animal models. The systematic search was conducted across four electronic databases: Ovid, Web of Science, PubMed and EBSCOHOST, based on PRISMA-ScR guidelines until November 2021. The inclusion criteria were: (i) animal models undergoing alveolar bone regenerative surgery, (ii) the intervention to regenerate or augment bone using 3D-printed hydroxyapatite or other calcium phosphate scaffolds and (iii) histological and microcomputed tomographic analyses of new bone formation and biological properties of 3D-printed hydroxyapatite or calcium phosphates. A total of ten studies were included in the review. All the studies showed promising results on new bone formation without any inflammatory reactions, regardless of the animal species. In conclusion, hydroxyapatite and tricalcium phosphates are feasible materials for 3D-printed scaffolds for alveolar bone regeneration and demonstrated bone regenerative potential in the oral cavity. However, further research is warranted to determine the scaffold material which mimics the gold standard of care for bone regeneration in the load-bearing areas, including the masticatory load of the oral cavity.
Collapse
Affiliation(s)
- Nurulhuda Mohd
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Masfueh Razali
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Mariyam Jameelah Ghazali
- Department of Mechanical & Manufacturing Engineering, Faculty of Engineering & Built Environment, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia;
| | - Noor Hayaty Abu Kasim
- Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| |
Collapse
|
36
|
Kern C, Jamous R, El Khassawna T, Rohnke M. Characterisation of Sr 2+ mobility in osteoporotic rat bone marrow by cryo-ToF-SIMS and cryo-OrbiSIMS. Analyst 2022; 147:4141-4157. [DOI: 10.1039/d2an00913g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mass spectrometric imaging approach for ex vivo monitoring of drug transport in bone sections. Cryo-ToF-SIMS depth profiling and high-resolution imaging as well as OrbiSIMS analysis revealed inhomogeneous Sr2+ transport in rat bone marrow.
Collapse
Affiliation(s)
- Christine Kern
- Institute of Physical Chemistry, Justus Liebig University Giessen, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Reem Jamous
- Experimental Trauma Surgery, Faculty of Medicine, Justus Liebig University Giessen, Aulweg 128, 35392 Giessen, Germany
| | - Thaqif El Khassawna
- Experimental Trauma Surgery, Faculty of Medicine, Justus Liebig University Giessen, Aulweg 128, 35392 Giessen, Germany
| | - Marcus Rohnke
- Institute of Physical Chemistry, Justus Liebig University Giessen, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| |
Collapse
|
37
|
Ekeuku SO, Chin KY, Qian J, Qu H, Wang Y, Ramli ESM, Wong SK, Noor MMM, Ima-Nirwana S. Normalisation of High Bone Remodelling due to Oestrogen Deficiency by Traditional Chinese Formulation Kang Shuai Lao Pian in Ovariectomised Rats. Int J Med Sci 2022; 19:1648-1659. [PMID: 36237992 PMCID: PMC9553853 DOI: 10.7150/ijms.75915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/09/2022] [Indexed: 11/05/2022] Open
Abstract
Postmenopausal osteoporosis transpires due to excessive osteoclastic bone resorption and insufficient osteoblastic bone formation in the presence of oestrogen insufficiency. Kang Shuai Lao Pian (KSLP) is a red ginseng-based traditional Chinese medicine known for its anti-ageing properties. However, studies on its effect on bone loss are lacking. Thus, the current study examined the skeletal protective effects of KSLP in an ovariectomised rodent bone loss model. Three-month-old female Sprague Dawley rats (n=42) were randomised into baseline, sham and ovariectomised (OVX) groups. The OVX rats were supplemented with low- (KSLP-L; 0.15 g/kg), medium- (KSLP-M; 0.30 g/kg), high-dose KSLP (KSLP-H; 0.45 g/kg) or calcium carbonate (1% w/v). The daily supplementation of KSLP was performed via oral gavage for eight weeks. Gavage stress was stimulated in the ovariectomised control with distilled water. The rats were euthanised at the end of the study. Whole-body and femoral bone mineral content and density scans were performed at baseline and every four weeks. Blood samples were obtained for the determination of bone remodelling markers. Histomorphometry and biomechanical strength testing were performed on femurs and tibias. High bone remodelling typically due to oestrogen deficiency, indicated by the elevated bone formation and resorption markers, osteoclast surface, single-labelled surface and mineralising surface/bone surface ratio, was observed in the untreated OVX rats. Whole-body BMD adjusted to body weight and Young's modulus decreased significantly in the untreated OVX rats. High-dose KSLP supplementation counteracted these degenerative changes. In conclusion, KSLP improves bone health by normalising bone remodelling, thereby preventing bone loss and decreased bone strength caused by oestrogen deficiency. Its anti-osteoporosis effects should be validated in patients with postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Sophia Ogechi Ekeuku
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Haibin Qu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Elvy Suhana Mohd Ramli
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Mustazil Mohd Noor
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Foessl I, Bassett JHD, Bjørnerem Å, Busse B, Calado Â, Chavassieux P, Christou M, Douni E, Fiedler IAK, Fonseca JE, Hassler E, Högler W, Kague E, Karasik D, Khashayar P, Langdahl BL, Leitch VD, Lopes P, Markozannes G, McGuigan FEA, Medina-Gomez C, Ntzani E, Oei L, Ohlsson C, Szulc P, Tobias JH, Trajanoska K, Tuzun Ş, Valjevac A, van Rietbergen B, Williams GR, Zekic T, Rivadeneira F, Obermayer-Pietsch B. Bone Phenotyping Approaches in Human, Mice and Zebrafish - Expert Overview of the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal traits TranslatiOnal NEtwork"). Front Endocrinol (Lausanne) 2021; 12:720728. [PMID: 34925226 PMCID: PMC8672201 DOI: 10.3389/fendo.2021.720728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
A synoptic overview of scientific methods applied in bone and associated research fields across species has yet to be published. Experts from the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal Traits translational Network") Working Group 2 present an overview of the routine techniques as well as clinical and research approaches employed to characterize bone phenotypes in humans and selected animal models (mice and zebrafish) of health and disease. The goal is consolidation of knowledge and a map for future research. This expert paper provides a comprehensive overview of state-of-the-art technologies to investigate bone properties in humans and animals - including their strengths and weaknesses. New research methodologies are outlined and future strategies are discussed to combine phenotypic with rapidly developing -omics data in order to advance musculoskeletal research and move towards "personalised medicine".
Collapse
Affiliation(s)
- Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| | - J. H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Åshild Bjørnerem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Norwegian Research Centre for Women’s Health, Oslo University Hospital, Oslo, Norway
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Ângelo Calado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | | | - Maria Christou
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Eleni Douni
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Imke A. K. Fiedler
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Eva Hassler
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University Graz, Graz, Austria
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Erika Kague
- The School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, United Kingdom
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Patricia Khashayar
- Center for Microsystems Technology, Imec and Ghent University, Ghent, Belgium
| | - Bente L. Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Victoria D. Leitch
- Innovative Manufacturing Cooperative Research Centre, Royal Melbourne Institute of Technology, School of Engineering, Carlton, VIC, Australia
| | - Philippe Lopes
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Georgios Markozannes
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | | | | | - Evangelia Ntzani
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
- Department of Health Services, Policy and Practice, Center for Research Synthesis in Health, School of Public Health, Brown University, Providence, RI, United States
| | - Ling Oei
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pawel Szulc
- INSERM UMR 1033, University of Lyon, Lyon, France
| | - Jonathan H. Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, Bristol, University of Bristol, Bristol, United Kingdom
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Şansın Tuzun
- Physical Medicine & Rehabilitation Department, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Amina Valjevac
- Department of Human Physiology, School of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Bert van Rietbergen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Tatjana Zekic
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | | | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| |
Collapse
|
39
|
Medeiros Savi F, Mieszczanek P, Revert S, Wille ML, Bray LJ. A New Automated Histomorphometric MATLAB Algorithm for Immunohistochemistry Analysis Using Whole Slide Imaging. Tissue Eng Part C Methods 2021; 26:462-474. [PMID: 32729382 DOI: 10.1089/ten.tec.2020.0153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The use of animal models along with the employment of advanced and sophisticated stereological methods for assessing bone quality combined with the use of statistical methods to evaluate the effectiveness of bone therapies has made it possible to investigate the pathways that regulate bone responses to medical devices. Image analysis of histomorphometric measurements remains a time-consuming task, as the image analysis software currently available does not allow for automated image segmentation. Such a feature is usually obtained by machine learning and with software platforms that provide image-processing tools such as MATLAB. In this study, we introduce a new MATLAB algorithm to quantify immunohistochemically stained critical-sized bone defect samples and compare the results with the commonly available Aperio Image Scope Positive Pixel Count (PPC) algorithm. Bland and Altman analysis and Pearson correlation showed that the measurements acquired with the new MATLAB algorithm were in excellent agreement with the measurements obtained with the Aperio PPC algorithm, and no significant differences were found within the histomorphometric measurements. The ability to segment whole slide images, as well as defining the size and the number of regions of interest to be quantified, makes this MATLAB algorithm a potential histomorphometric tool for obtaining more objective, precise, and reproducible quantitative assessments of entire critical-sized bone defect image data sets in an efficient and manageable workflow.
Collapse
Affiliation(s)
- Flavia Medeiros Savi
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,School of Mechanical, Medical and Process Engineering, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Pawel Mieszczanek
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Sophia Revert
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Marie-Luise Wille
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,School of Mechanical, Medical and Process Engineering, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland, Australia.,ARC ITTC for Multiscale 3D Imaging, Modelling, and Manufacturing, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Laura Jane Bray
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,School of Mechanical, Medical and Process Engineering, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
40
|
Emmanuel T, Brüel A, Thomsen JS, Steiniche T, Brent MB. Artificial intelligence-assisted identification and quantification of osteoclasts. MethodsX 2021; 8:101272. [PMID: 34434793 PMCID: PMC8374260 DOI: 10.1016/j.mex.2021.101272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/15/2021] [Accepted: 02/11/2021] [Indexed: 11/26/2022] Open
Abstract
Quantification of osteoclasts to assess bone resorption is a time-consuming and tedious process. Since the inception of bone histomorphometry and manual counting of osteoclasts using bright-field microscopy, several approaches have been proposed to accelerate the counting process using both free and commercially available software. However, most of the present alternatives depend on manual or semi-automatic color segmentation and do not take advantage of artificial intelligence (AI). The present study directly compare estimates of osteoclast-covered surfaces (Oc.S/BS) obtained by the conventional manual method using a bright-field microscope to that obtained by a new AI-assisted method. We present a detailed step-by-step guide for the AI-based method. Tibiae from Wistar rats were either enzymatically stained for TRAP or immunostained for cathepsin K to identify osteoclasts. We found that estimation of Oc.S/BS by the new AI-assisted method was considerably less time-consuming, while still providing similar results to the conventional manual method. In addition, the retrainable AI-module used in the present study allows for fully automated overnight batch processing of multiple annotated sections.Bone histomorphometry AI-assisted osteoclast identification TRAP and cathepsin K
Collapse
Affiliation(s)
- Thomas Emmanuel
- Department of Dermatology, Aarhus University Hospital, Denmark
| | | | | | | | | |
Collapse
|
41
|
Mostafa AA, Mahmoud AA, Hamid MAA, Basha M, El-Okaily MS, Abdelkhalek AFA, El-Anwar MI, El Moshy S, Gibaly A, Hassan EA. An in vitro / in vivo release test of risedronate drug loaded nano-bioactive glass composite scaffolds. Int J Pharm 2021; 607:120989. [PMID: 34389417 DOI: 10.1016/j.ijpharm.2021.120989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 01/06/2023]
Abstract
Three-dimensional (3D) matrices scaffolds play a noteworthy role in promoting cell generation and propagation. In this study, scaffolds prepared from chitosan/polyvinyl alcohol loaded with/without an osteoporotic drug (risedronate) and nano-bioactive glass (nBG) have been developed to promote healing of bone defects. The scaffolds were characterized by scanning electron microscopy (SEM), porosity test as well as mechanical strength. The pattern of drug release and ability to promote the proliferation of Saos-2osteosarcoma cells had also been reported. Osteogenic potential of the scaffolds was evaluated by testing their effect on healing critical-sized dog's mandibular bone defects. Increasing chitosan and nBG in the porous scaffolds induced decrease in drug release, increased the scaffold's strength and supported their cell proliferation, alkaline phosphatase (ALP) activities, as well as increased calcium deposition. Histological and histomorphometric results demonstrated newly formed bone trabeculae inside critical-sized mandibular defects when treated with scaffolds. Trabecular thickness, bone volume/tissue volume and the percentage of mature collagen fibers increased in groups treated with scaffolds loaded with 10% nBG and risedronate or loaded with 30% nBG with/without risedronate compared with those treated with non-loaded scaffolds and empty control groups. These findings confirmed the potential osteogenic activity of chitosan/polyvinyl alcohol-based scaffolds loaded with risedronate and nBG.
Collapse
Affiliation(s)
- Amany A Mostafa
- Nanomedicine & Tissue Engineering Lab., Medical Research Center of Excellence (MRCE), National Research Centre, Cairo, Egypt; Refractories, Ceramics & Building Materials Department (Biomaterials group), National Research Centre, Cairo, Egypt.
| | - Azza A Mahmoud
- Nanomedicine & Tissue Engineering Lab., Medical Research Center of Excellence (MRCE), National Research Centre, Cairo, Egypt; Department of Pharmaceutical Technology, Pharmaceutical and Drug Industries Research Division, National Research Centre, Cairo, Egypt; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Mohamed A Abdel Hamid
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mona Basha
- Department of Pharmaceutical Technology, Pharmaceutical and Drug Industries Research Division, National Research Centre, Cairo, Egypt
| | - Mohamed S El-Okaily
- Nanomedicine & Tissue Engineering Lab., Medical Research Center of Excellence (MRCE), National Research Centre, Cairo, Egypt; Refractories, Ceramics & Building Materials Department (Biomaterials group), National Research Centre, Cairo, Egypt
| | - Abdel Fattah A Abdelkhalek
- Department of Microbiology of Supplementary General Science, Faculty of Oral & Dental Medicine, Future University in Egypt, Cairo, Egypt
| | - Mohamed I El-Anwar
- Department of Mechanical Engineering, National Research Centre, Cairo, Egypt
| | - Sara El Moshy
- Department of Oral Biology, Faculty of Dentistry, Cairo University, Cairo, Egypt
| | - Amr Gibaly
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Beni-Suef University, Beni-Suef, Egypt
| | - Elham A Hassan
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
42
|
Tiwari AK, Goyal A, Prasad J. Modeling cortical bone adaptation using strain gradients. Proc Inst Mech Eng H 2021; 235:636-654. [PMID: 33754910 DOI: 10.1177/09544119211000228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cyclic and low-magnitude loading promotes osteogenesis (i.e. new bone formation). Normal strain, strain energy density and fatigue damage accumulation are typically considered as osteogenic stimuli in computer models to predict site-specific new bone formation. These models however had limited success in explaining osteogenesis near the sites of minimal normal strain, for example, neutral axis of bending. Other stimuli such as fluid motion or strain gradient also stimulate bone formation. In silico studies modeled the new bone formation as a function of fluid motion, however, computation of fluid motion involves complex mathematical calculations. Strain gradients drive fluid flow and thus can also be established as the stimulus. Osteogenic potential of strain gradients is however not well established. The present study establishes strain gradients as osteogenic stimuli. Bending-induced strain gradients are computed at cortical bone cross-sections reported in animal loading in vivo studies. Correlation analysis between strain gradients and site of osteogenesis is analyzed. In silico model is also developed to test the osteogenic potential of strain gradients. The model closely predicts in vivo new bone distribution as a function of strain gradients. The outcome establishes strain gradient as computationally easy and robust stimuli to predict site-specific osteogenesis. The present study may be useful in the development of biomechanical approaches to mitigate bone loss.
Collapse
Affiliation(s)
- Abhishek Kumar Tiwari
- Department of Applied Mechanics, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, India
| | - Ajay Goyal
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Jitendra Prasad
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| |
Collapse
|
43
|
Shapiro F, Maguire K, Swami S, Zhu H, Flynn E, Wang J, Wu JY. Histopathology of osteogenesis imperfecta bone. Supramolecular assessment of cells and matrices in the context of woven and lamellar bone formation using light, polarization and ultrastructural microscopy. Bone Rep 2021; 14:100734. [PMID: 33665234 PMCID: PMC7898004 DOI: 10.1016/j.bonr.2020.100734] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
Diaphyseal long bone cortical tissue from 30 patients with lethal perinatal Sillence II and progressively deforming Sillence III osteogenesis imperfecta (OI) has been studied at multiple levels of structural resolution. Interpretation in the context of woven to lamellar bone formation by mesenchymal osteoblasts (MOBLs) and surface osteoblasts (SOBLs) respectively demonstrates lamellar on woven bone synthesis as an obligate self-assembly mechanism and bone synthesis following the normal developmental pattern but showing variable delay in maturation caused by structurally abnormal or insufficient amounts of collagen matrix. The more severe the variant of OI is, the greater the persistence of woven bone and the more immature the structural pattern; the pattern shifts to a structurally stronger lamellar arrangement once a threshold accumulation for an adequate scaffold of woven bone has been reached. Woven bone alone characterizes lethal perinatal variants; variable amounts of woven and lamellar bone occur in progressively deforming variants; and lamellar bone increasingly forms rudimentary and then partially compacted osteons not reaching full compaction. At differing levels of microscopic resolution: lamellar bone is characterized by short, obliquely oriented lamellae with a mosaic appearance in progressively deforming forms; polarization defines tissue conformations and localizes initiation of lamellar formation; ultrastructure of bone forming cells shows markedly dilated rough endoplasmic reticulum (RER) and prominent Golgi bodies with disorganized cisternae and swollen dispersed tubules and vesicles, structural indications of storage disorder/stress responses and mitochondrial swelling in cells with massively dilated RER indicating apoptosis; ultrastructural matrix assessments in woven bone show randomly oriented individual fibrils but also short pericellular bundles of parallel oriented fibrils positioned obliquely and oriented randomly to one another and in lamellar bone show unidirectional fibrils that deviate at slight angles to adjacent bundles and obliquely oriented fibril groups consistent with twisted plywood fibril organization. Histomorphometric indices, designed specifically to document woven and lamellar conformations in normal and OI bone, establish ratios for: i) cell area/total area X 100 indicating the percentage of an area occupied by cells (cellularity index) and ii) total area/number of cells (pericellular matrix domains). Woven bone is more cellular than lamellar bone and OI bone is more cellular than normal bone, but these findings occur in a highly specific fashion with values (high to low) encompassing OI woven, normal woven, OI lamellar and normal lamellar conformations. Conversely, for the total area/number of cells ratio, pericellular matrix accumulations in OI woven are smallest and normal lamellar largest. Since genotype-phenotype correlation is not definitive, interposing histologic/structural analysis allowing for a genotype-histopathologic-phenotype correlation will greatly enhance understanding and clinical management of OI.
Collapse
Affiliation(s)
- Frederic Shapiro
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kathleen Maguire
- Division of Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Srilatha Swami
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Palo Alto, CA, USA
| | - Hui Zhu
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Palo Alto, CA, USA
| | - Evelyn Flynn
- Orthopaedic Research Laboratory, Boston Children's Hospital, Boston, MA, USA
| | - Jamie Wang
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Palo Alto, CA, USA
| | - Joy Y Wu
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
44
|
Rowe DW, Hong SH, Zhang C, Shin DG, Adams DJ, Youngstrom DW, Chen L, Wu Z, Zhou Y, Maye P. Skeletal screening IMPC/KOMP using μCT and computer automated cryohistology: Application to the Efna4 KO mouse line. Bone 2021; 144:115688. [PMID: 33065355 DOI: 10.1016/j.bone.2020.115688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023]
Abstract
The IMPC/KOMP program provides the opportunity to screen mice harboring well defined gene-inactivation mutations in a uniform genetic background. The program performs a global tissue phenotyping survey that includes skeletal x-rays and bone density measurements. Because of the relative insensitivity of the two screening tests for detecting variance in bone architecture, we initiated a secondary screen based on μCT and a cryohistolomorphological workflow that was performed on the femur and vertebral compartments on 220 randomly selected knockouts (KOs) and 36 control bone samples over a 2 1/2 year collection period provided by one of the production/phenotyping centers. The performance of the screening protocol was designed to balance throughput and cost versus sensitivity and informativeness such that the output would be of value to the skeletal biology community. Here we report the reliability of this screening protocol to establish criteria for control skeletal variance at the architectural, dynamic and cellular histomorphometric level. Unexpected properties of the control population include unusually high variance in BV/TV in male femurs and greater bone formation and bone turnover rates in the female femur and vertebral trabeculae bone compartments. However, the manner for maintaining bone formation differed between these two bone sites. The vertebral compartment relies on maintaining a greater number of bone forming surfaces while the femoral compartment utilized more matrix production per cell. The comparison of the architectural properties obtained by μCT and histomorphology revealed significant differences in values for BV/TV, Tb.Th and Tb.N which is attributable to sampling density of the two methods. However, as a screening tool, expressing the ratio of KO to the control line as obtained by either method was remarkably similar. It identified KOs with significant variance which led to a more detailed histological analysis. Our findings are exemplified by the Efna4 KO, in which a high BV/TV was identified by μCT and confirmed by histomorphometry in the femur but not in the vertebral compartment. Dynamic labeling showed a marked increase in BFR which was attributable to increased labeling surfaces. Cellular analysis confirmed partitioning of osteoblast to labeling surfaces and a marked decrease in osteoclastic activity on both labeling and quiescent surfaces. This pattern of increased bone modeling would not be expected based on prior studies of the Ephrin-Ephrin receptor signaling pathways between osteoblasts and osteoclasts. Overall, our findings underscore why unbiased screening is needed because it can reveal unknown or unanticipated genes that impact skeletal variation.
Collapse
Affiliation(s)
- David W Rowe
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America.
| | - Seung-Hyun Hong
- Computer Science & Engineering, School of Engineering, University of Connecticut, Storrs, CT 06269, United States of America
| | - Caibin Zhang
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Dong-Guk Shin
- Computer Science & Engineering, School of Engineering, University of Connecticut, Storrs, CT 06269, United States of America
| | - Douglas J Adams
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, School of Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Li Chen
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Zhihua Wu
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Yueying Zhou
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Peter Maye
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| |
Collapse
|
45
|
Rahmati M, Stötzel S, Khassawna TE, Iskhahova K, Florian Wieland DC, Zeller Plumhoff B, Haugen HJ. Early osteoimmunomodulatory effects of magnesium-calcium-zinc alloys. J Tissue Eng 2021; 12:20417314211047100. [PMID: 34589198 PMCID: PMC8474317 DOI: 10.1177/20417314211047100] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/01/2021] [Indexed: 01/04/2023] Open
Abstract
Today, substantial attention is given to biomaterial strategies for bone regeneration, and among them, there is a growing interest in using immunomodulatory biomaterials. The ability of a biomaterial to induce neo vascularization and macrophage polarization is a major factor in defining its success. Magnesium (Mg)-based degradable alloys have attracted significant attention for bone regeneration owing to their biodegradability and potential for avoiding secondary removal surgeries. However, there is insufficient evidence in the literature regarding the early inflammatory responses to these alloys in vivo. In this study, we investigated the early body responses to Mg-0.45wt%Zn-0.45wt%Ca pin-shaped alloy (known as ZX00 alloy) in rat femora 2, 5, and 10 days after implantation. We used 3D micro computed tomography (µCT), histological, immunohistochemical, histomorphometrical, and small angle X-ray scattering (SAXS) analyses to study new bone formation, early macrophage polarization, neo vascularization, and bone quality at the implant bone interface. The expression of macrophage type 2 biological markers increased significantly after 10 days of Mg alloy implantation, indicating its potential in stimulating macrophage polarization. Our biomineralization results using µCT as well as histological stained sections did not indicate any statistically significant differences between different time points for both groups. The activity of alkaline phosphatase (ALP) and Runt-related transcription factor 2 (Runx 2) biological markers decreased significantly for Mg group, indicating less osteoblast activity. Generally, our results supported the potential of ZX00 alloy to enhance the expression of macrophage polarization in vivo; however, we could not observe any statistically significant changes regarding biomineralization.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Biomaterials, Institute
for Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Sabine Stötzel
- Experimental Trauma Surgery,
Justus-Liebig University Giessen, Giessen, Germany
| | - Thaqif El Khassawna
- Experimental Trauma Surgery,
Justus-Liebig University Giessen, Giessen, Germany
- Faculty of Health Sciences, University
of Applied Sciences, Giessen, Germany
| | - Kamila Iskhahova
- Institute of Metallic Biomaterials,
Helmholtz-Zentrum Hereon, Geesthacht, Germany
| | - DC Florian Wieland
- Institute of Metallic Biomaterials,
Helmholtz-Zentrum Hereon, Geesthacht, Germany
| | | | - Håvard Jostein Haugen
- Department of Biomaterials, Institute
for Clinical Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
46
|
A novel RGB-trichrome staining method for routine histological analysis of musculoskeletal tissues. Sci Rep 2020; 10:16659. [PMID: 33028938 PMCID: PMC7541469 DOI: 10.1038/s41598-020-74031-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Morphometry and histology are essential approaches for investigation and diagnosis of musculo-skeletal disorders. Despite the advent of revolutionary methods of image analysis and high resolution three-dimensional imaging technology, basic conventional light microscopy still provides an incisive overview of the structure and tissue dynamics of the musculoskeletal system. This is crucial to both preclinical and clinical research, since several clinically relevant processes, such as bone repair, osteoarthritis, and metabolic bone diseases, display distinct, if not pathognomonic, histological features. Due to the particular characteristics of the skeletal tissues (i.e., the existence of mineralized extracellular matrices), a large number of staining methods applicable to either decalcified or undecalcified tissues are available. However, it is usually the case that several staining methods need to be sequentially applied in order to achieve the different endpoints required to fully assess skeletal tissue structure and dynamics, and to allow morphometric quantification. We describe herein a novel staining method, the RGB trichrome, amenable for application to decalcified, paraffin embedded human musculoskeletal tissues. The acronym RGB corresponds to the three primary dyes used: picrosirius Red, fast Green, and alcian Blue. Although these individual pigments are commonly used either isolated, in binary combinations, or as part of more complex polychrome staining methods, when merged in the RGB trichrome staining produce high-quality/high-contrast images, permitting not only clear identification of different tissues (i.e., the different types of cartilage, bone and fibrous connective tissue), but also discrimination between calcified and uncalcified bone and cartilage, as well as an unexpected diversity of shades of color, while displaying singular properties among polychrome staining methods, such as the unveiling of the bone osteocyte dendritic/canalicular network. Hence, we propose the RGB trichrome as simple but highly-reliable tool for the preclinical and clinical study of the musculoskeletal system.
Collapse
|
47
|
Portier H, Benaitreau D, Pallu S. Does Physical Exercise Always Improve Bone Quality in Rats? Life (Basel) 2020; 10:life10100217. [PMID: 32977460 PMCID: PMC7598192 DOI: 10.3390/life10100217] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022] Open
Abstract
For decades, the osteogenic effect from different physical activities on bone in rodents remained uncertain. This literature review presents for the first time the effects on five exercise models (treadmill running, wheel running, swimming, resistance training and vibration modes) in three different experimental rat groups (males, females, osteopenic) on bone quality. The bone parameters presented are bone mineral density, micro-architectural and mechanical properties, and osteoblast/osteocyte and osteoclast parameters. This review shows that physical activities have a positive effect (65% of the results) on bone status, but we clearly observed a difference amongst the different protocols. Even if treadmill running is the most used protocol, the resistance training constitutes the first exercise model in term of osteogenic effects (87% of the whole results obtained on this model). The less osteogenic model is the vibration mode procedure (31%). It clearly appears that the gender plays a role on the bone response to swimming and wheel running exercises. Besides, we did not observe negative results in the osteopenic population with impact training, wheel running and vibration activities. Moreover, about osteoblast/osteocyte parameters, we conclude that high impact and resistance exercise (such jumps and tower climbing) seems to increase bone formation more than running or aerobic exercise. Among the different protocols, literature has shown that the treadmill running procedure mainly induces osteogenic effects on the viability of the osteocyte lineage in both males and females or ovariectomized rats; running in voluntary wheels contributes to a negative effect on bone metabolism in older male models; whole-body vertical vibration is not an osteogenic exercise in female and ovariectomized rats; whereas swimming provides controversial results in female models. For osteoclast parameters only, running in a voluntary wheel for old males, the treadmill running program at high intensity in ovariectomized rats, and the swimming program in a specific ovariectomy condition have detrimental consequences.
Collapse
Affiliation(s)
- Hugues Portier
- Laboratoire de Biologie Bioingénierie et Bioimagerie Ostéo-Articulaire (B3OA), Université Paris, UMR CNRS 7052, INSERM U1273, 10 Av de Verdun, 75010 Paris, France;
- Collegium Science & Technique, 2 allée du château, Université d’Orléans. 45100 Orléans, France;
- Correspondence: ; Tel.: +33-782-309-433
| | - Delphine Benaitreau
- Collegium Science & Technique, 2 allée du château, Université d’Orléans. 45100 Orléans, France;
| | - Stéphane Pallu
- Laboratoire de Biologie Bioingénierie et Bioimagerie Ostéo-Articulaire (B3OA), Université Paris, UMR CNRS 7052, INSERM U1273, 10 Av de Verdun, 75010 Paris, France;
- Collegium Science & Technique, 2 allée du château, Université d’Orléans. 45100 Orléans, France;
| |
Collapse
|
48
|
Ikeda T, Okano S, Hashimoto N, Kimura K, Kudo K, Tsutsumi R, Sasaki S, Kawasaki J, Miyashita Y, Wada H. Histomorphological investigation of intrahepatic connective tissue for surgical anatomy based on modern computer imaging analysis. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2020; 28:76-85. [PMID: 32697892 PMCID: PMC7891672 DOI: 10.1002/jhbp.753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/09/2020] [Accepted: 04/22/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND/PURPOSE Computer-assisted tissue imaging and analytical techniques were used to clarify the histomorphological structure of hepatic connective tissue as a practical guide for surgeons. METHODS Approximately 5000 histological slides were prepared from liver specimens of five autopsied patients. Three-dimensional (3D) reconstruction was performed and subjected to computer imaging analysis. Scanning electron microscopy was also performed on the liver specimens. RESULTS The 3D reconstructed images revealed the running form of the vasculature and the relationship between the hepatic lobule and connective tissue. The hepatic capsule or portal pedicle was consistently located at the periphery of the hepatic lobules. An artificial intelligence random forest approach clearly segmented hepatic cells, type I collagen (CF), type III collagen (RF), and other cells. The hepatic lobule, portal region, and hepatic capsule were significantly distinguished based on CF and RF occupancy. The capsule directly covering the liver lobule with an RF concentration up to 87% was provisionally named the proper hepatic capsule. The existence of a proper hepatic ligament with distinct occupation rates of CF and RF was also suggested. CONCLUSIONS The identified proper hepatic capsule and ligament can be important markers for demarcating the dissecting layer during surgical procedures.
Collapse
Affiliation(s)
- Tetsuo Ikeda
- Department of Integration of Advanced Medicine and Innovative Technology, Kyushu University Hospital, Fukuoka, Japan.,Department of Endoscopy and Endoscopic Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Shinji Okano
- Department of Pathology, Fukuoka Dental College, Fukuoka, Japan
| | - Naotaka Hashimoto
- Department of Endoscopy and Endoscopic Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Koichi Kimura
- Department of Endoscopy and Endoscopic Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Kensuke Kudo
- Department of Endoscopy and Endoscopic Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Ryosuke Tsutsumi
- Department of Endoscopy and Endoscopic Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Shun Sasaki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junji Kawasaki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yu Miyashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroya Wada
- Department of Endoscopy and Endoscopic Surgery, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
49
|
Abstract
The present work focuses on the application of time-of-flight secondary ion mass spectrometry (ToF-SIMS) in osteoporotic bone research. In order to demonstrate the benefit, the authors present concrete application examples of ToF-SIMS in three different areas of bone research. ToF-SIMS as a mass spectrometric imaging technique allows simultaneous visualization of mineralized and nonmineralized bone tissue as well as implanted biomaterials and bone implant interphases. In the first example, the authors show that it is possible to study the incorporation and distribution of different components released from bone filler materials into bone with a single mass spectrometric measurement. This not only enables imaging of nonstained bone cross sections but also provides further insights beyond histologically obtained information. Furthermore, they successfully identified several mass fragments as markers for newly formed cartilage tissue and growth joint in bone. Different modes of ToF-SIMS as well as different SIMS instruments (IONTOF's TOF.SIMS 5 and M6 Hybrid SIMS, Ionoptika's J105) were used to identify these mass signals and highlight the high versatility of this method. In the third part, bone structure of cortical rat bone was investigated from bone sections embedded in technovit (polymethyl methacrylate, PMMA) and compared to cryosections. In cortical bone, they were able to image different morphological features, e.g., concentric arrangement of collagen fibers in so-called osteons as well as Haversian canals and osteocytes. In summary, the study provides examples of application and shows the strength of ToF-SIMS as a promising analytical method in the field of osteoporotic bone research.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Artificial intelligence tools have found new applications in medical diagnosis. These tools have the potential to capture underlying trends and patterns, otherwise impossible with previous modeling capabilities. Machine learning and deep learning models have found a role in osteoporosis, both to model the risk of fragility fracture, and to help with the identification and segmentation of images. RECENT FINDINGS Here we survey the latest research in the artificial intelligence application to the prediction of osteoporosis that has been published between January 2017 and March 2019. Around half of the articles that are covered here predict (by classification or regression) an indicator of osteoporosis, such as bone mass or fragility fractures; the other half of studies use tools for automatic segmentation of the images of patients with or at risk of osteoporosis. The data for these studies include diverse signal sources: acoustics, MRI, CT, and of course, X-rays. SUMMARY New methods for automatic image segmentation, and prediction of fracture risk show promising clinical value. Though these recent developments have had a successful initial application to osteoporosis research, their development is still under improvement, such as accounting for positive/negative class bias. We urge care when reporting accuracy metrics, and when comparing such metrics between different studies.
Collapse
|