1
|
Steyn M, Brits D, Botha D, Holland S. Violence against children: A review of cases at a forensic anthropology unit, Johannesburg, South Africa. J Forensic Leg Med 2024; 101:102623. [PMID: 38043241 DOI: 10.1016/j.jflm.2023.102623] [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: 09/29/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/05/2023]
Abstract
South Africa is experiencing a scourge of violence against women and children, with alarming levels of violence, and as such, juvenile cases are sometimes encountered in a forensic anthropological setting. The aim of this research was to assess the cases of juveniles (under 20 years) presenting at the Human Variation and Identification Research Unit (HVIRU) for a 6-year period (2016-2022), in order to assess patterns and types of cases referred. A total of 19 cases were assessed, of which 10 were 16-20 years old, 3 between the ages of 5 and 15 years and 6 less than 5 years old at the time of death. Of the 14 children with known sex, 12 were female with features suggesting that they fell prey to sexual violence. Many of the individuals were either known (and referred for trauma analysis) or identified following investigation. Of the 19 juveniles, 11 (58 %) had evidence of perimortem trauma (sharp and/or blunt), attesting to the violent nature of their deaths. Two cases had both sharp and blunt force trauma, of which one is a possible case of dismemberment. The remains of two individuals showed signs of perimortem burning, which may or may not be related to the cause of death. Three individuals had signs of antemortem (healed) trauma, which may suggest a longer period of abuse. Five of the individuals showed signs of disease - two had cribra orbitalia, while two others had various porous lesions indicative of chronic disease or malnutrition. One individual had advanced osteomyelitis, suggesting a natural cause of death. Some of these cases had unusual trauma and pathology, highlighting the contribution of forensic anthropologists.
Collapse
Affiliation(s)
- M Steyn
- Human Variation and Identification Research Unit, School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, South Africa.
| | - D Brits
- Human Variation and Identification Research Unit, School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - D Botha
- Human Variation and Identification Research Unit, School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - S Holland
- Department of Forensic Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| |
Collapse
|
2
|
Clement ND, Gaston MS, Simpson AH. Fractures in elderly mice demonstrate delayed ossification of the soft callus: a cellular and radiographic study. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2023; 33:977-985. [PMID: 35239001 PMCID: PMC10125932 DOI: 10.1007/s00590-022-03235-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/14/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The aim of this study was to assess the cellular age-related changes in fracture repair and relate these to the observed radiographic assessments at differing time points. METHODS Transverse traumatic tibial diaphyseal fractures were created in 12-14 weeks old (young n = 16) and 18 months old (elderly n = 20) in Balb/C wild mice. Fracture calluses were harvested at five time points from 1 to 35 days post fracture for histomorphometry (percent of cartilage and bone), radiographic analysis (total callus volume, callus index, and relative bone mineral content). RESULTS The elderly mice produced an equal amount of cartilage when compared to young mice (p > 0.08). However, by day 21 there was a significantly greater percentage of bone at the fracture site in the young group (mean percentage 50% versus 11%, p < 0.001). It was not until day 35 when the elderly group produced a similar amount of bone compared to the young group at 21 days (50% versus 53%, non-significant (ns)). The callus area and callus index on radiographic assessment was not significantly different between young and elderly groups at any time point. Relative bone mineral content was significantly greater in the young group at 14 days (545.7 versus -120.2, p < 0.001) and 21 days (888.7 versus 451.0, p < 0.001) when compared to the elderly group. It was not until day 35 when the elderly group produced a similar relative bone mineral content as the young group at 21 days (888.7 versus 921.8, ns). CONCLUSIONS Elderly mice demonstrated a delay in endochondral ossification which was associated with a decreased relative bone mineral content at the fracture site and may help assess these cellular changes in a clinical setting.
Collapse
Affiliation(s)
- N. D. Clement
- Department of Orthopaedics and Trauma, University of Edinburgh, Little France, Edinburgh, EH16 4SA UK
| | - M. S. Gaston
- Department of Orthopaedics and Trauma, University of Edinburgh, Little France, Edinburgh, EH16 4SA UK
| | - A. H. Simpson
- Department of Orthopaedics and Trauma, University of Edinburgh, Little France, Edinburgh, EH16 4SA UK
| |
Collapse
|
3
|
Gao H, Huang J, Wei Q, He C. Advances in Animal Models for Studying Bone Fracture Healing. Bioengineering (Basel) 2023; 10:bioengineering10020201. [PMID: 36829695 PMCID: PMC9952559 DOI: 10.3390/bioengineering10020201] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Fracture is a common traumatic injury that is mostly caused by traffic accidents, falls, and falls from height. Fracture healing is a long-term and complex process, and the mode of repair and rate of healing are influenced by a variety of factors. The prevention, treatment, and rehabilitation of fractures are issues that urgently need to be addressed. The preparation of the right animal model can accurately simulate the occurrence of fractures, identify and observe normal and abnormal healing processes, study disease mechanisms, and optimize and develop specific treatment methods. We summarize the current status of fracture healing research, the characteristics of different animal models and the modeling methods for different fracture types, analyze their advantages and disadvantages, and provide a reference basis for basic experimental fracture modeling.
Collapse
Affiliation(s)
- Hui Gao
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinming Huang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu 610041, China
- Correspondence: (Q.W.); (C.H.)
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu 610041, China
- Correspondence: (Q.W.); (C.H.)
| |
Collapse
|
4
|
van Trikt CH, Donders JCE, Klinger CE, Wellman DS, Helfet DL, Kloen P. Operative treatment of nonunions in the elderly: Clinical and radiographic outcomes in patients at minimum 75 years of age. BMC Geriatr 2022; 22:985. [PMID: 36539691 PMCID: PMC9764700 DOI: 10.1186/s12877-022-03670-8] [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: 03/29/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Limited information exists on nonunion treatment in the elderly. This retrospective study evaluates whether results of operative treatment of nonunion of the humerus or femur in patients aged ≥ 75 years are comparable to those in younger patients. METHODS We identified patients age ≥ 75 years with a nonunion of humerus or femur treated with open reduction and internal fixation. The Non-Union Scoring System was calculated. Complications, clinical outcome, and radiographic findings were assessed. Primary endpoint was nonunion healing. A literature review compared time to healing of humeral and femoral nonunion in younger populations. RESULTS We identified 45 patients treated for a nonunion of humerus or femur with > 12 months follow-up. Median age was 79 years (range 75-96). Median time to presentation was 12 months (range 4-127) after injury, median number of prior surgeries was 1 (range 0-4). Union rate was 100%, with median time to union 6 months (range 2-42). Six patients underwent revision for persistent nonunion and healed without further complications. CONCLUSIONS Using a protocol of debridement, alignment, compression, stable fixation, bone grafting and early motion, patients aged 75 years or older can reliably achieve healing when faced with a nonunion of the humerus or femur. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
- Clinton H. van Trikt
- grid.509540.d0000 0004 6880 3010Department of Orthopedic Surgery and Sports Medicine, Amsterdam University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Johanna C. E. Donders
- grid.509540.d0000 0004 6880 3010Department of Orthopedic Surgery and Sports Medicine, Amsterdam University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Craig E. Klinger
- grid.5386.8000000041936877XOrthopaedic Trauma Service, Hospital for Special Surgery and New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY USA
| | - David S. Wellman
- grid.260917.b0000 0001 0728 151XOrthopaedic Trauma Service, Westchester Medical Center, New York Medical College, Valhalla, NY USA
| | - David L. Helfet
- grid.5386.8000000041936877XOrthopaedic Trauma Service, Hospital for Special Surgery and New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY USA
| | - Peter Kloen
- grid.509540.d0000 0004 6880 3010Department of Orthopedic Surgery and Sports Medicine, Amsterdam University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Woloszyk A, Tuong ZK, Perez L, Aguilar L, Bankole AI, Evans CH, Glatt V. Fracture hematoma micro-architecture influences transcriptional profile and plays a crucial role in determining bone healing outcomes. BIOMATERIALS ADVANCES 2022; 139:213027. [PMID: 35882120 DOI: 10.1016/j.bioadv.2022.213027] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/27/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
The hematoma that forms between broken fragments of bone serves as a natural fibrin scaffold, and its removal from the defect site delays bone healing. The hypothesis of this study is that the microarchitectural and mechanical properties of the initially formed hematoma has a significant effect on the regulation of the biological process, which ultimately determines the outcome of bone healing. To mimic three healing conditions in the rat femur (normal, delayed, and non-healing bone defects), three different defect sizes of 0.5, 1.5, and 5.0 mm, are respectively used. The analysis of 3-day-old hematomas demonstrates clear differences in fibrin clot micro-architecture in terms of fiber diameter, fiber density, and porosity of the formed fibrin network, which result in different mechanical properties (stiffness) of the hematoma in each model. Those differences directly affect the biological processes involved. Specifically, RNA-sequencing reveals almost 700 differentially expressed genes between normally healing and non-healing defects, including significantly up-regulated essential osteogenic genes in normally healing defects, also differences in immune cell populations, activated osteogenic transcriptional regulators as well as potential novel marker genes. Most importantly, this study demonstrates that the healing outcome has already been determined during the hematoma phase of bone healing, three days post-surgery.
Collapse
Affiliation(s)
- Anna Woloszyk
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Zewen K Tuong
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba 4102, QLD, Australia; Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK; Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.
| | - Louis Perez
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Leonardo Aguilar
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Abraham I Bankole
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Christopher H Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester 55902, MN, USA.
| | - Vaida Glatt
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| |
Collapse
|
6
|
Ryan G, Magony R, Gortler H, Godbout C, Schemitsch EH, Nauth A. Systemically impaired fracture healing in small animal research: A review of fracture repair models. J Orthop Res 2021; 39:1359-1367. [PMID: 33580554 DOI: 10.1002/jor.25003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/09/2020] [Accepted: 02/10/2021] [Indexed: 02/04/2023]
Abstract
Fracture healing is a complex process requiring mechanical stability, an osteoconductive matrix, and osteoinductive and osteogenic biology. This intricate process is easily disrupted by various patient factors such as chronic disease and lifestyle. As the medical complexity and age of patients with fractures continue to increase, the importance of developing relevant experimental models is becoming paramount in preclinical research. The objective of this review is to describe the most common small animal models of systemically impaired fracture healing used in the orthopedic literature including osteoporosis, diabetes mellitus, smoking, alcohol use, obesity, and ageing. This review will provide orthopedic researchers with a summary of current models of systemically impaired fracture healing used in small animals and present an overview of the methods of induction for each condition.
Collapse
Affiliation(s)
- Gareth Ryan
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Richard Magony
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Hilary Gortler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Charles Godbout
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Emil H Schemitsch
- Department of Surgery, Division of Orthopaedic Surgery, University of Western Ontario, London, Ontario, Canada
| | - Aaron Nauth
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, Division of Orthopaedic Surgery, St. Michael's Hospital - Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Josephson AM, Leclerc K, Remark LH, Lopeź EM, Leucht P. Systemic NF-κB-mediated inflammation promotes an aging phenotype in skeletal stem/progenitor cells. Aging (Albany NY) 2021; 13:13421-13429. [PMID: 34035186 PMCID: PMC8202837 DOI: 10.18632/aging.203083] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/11/2021] [Indexed: 01/15/2023]
Abstract
Aging tissues undergo a progressive decline in regenerative potential. This decline in regenerative responsiveness has been attributed to changes in tissue-specific stem cells and their niches. In bone, aged skeletal stem/progenitor cell dysfunction is characterized by decreased frequency and impaired osteogenic differentiation potential. This aging phenotype ultimately results in compromised regenerative responsiveness to injury. The age-associated increase of inflammatory mediators, known as inflamm-aging, has been identified as the main culprit driving skeletal stem cell dysfunction. Here, we utilized a mouse model of parabiosis to decouple aging from inflammation. Using the Nfkb1-/- mouse as a model of inflamm-aging, we demonstrate that a shared systemic circulation between a wild-type and Nfkb1-/- mouse results in an aging phenotype of the wild-type skeletal stem and progenitor cells, shown by CFU-fs and osteogenic and adipogenic differentiation assays. Our findings demonstrate that exposure to an inflammatory secretome results in a phenotype similar to the one observed in aging.
Collapse
Affiliation(s)
- Anne Marie Josephson
- NYU Grossman School of Medicine, NYU Langone Orthopedics, New York, NY 10016, USA
| | - Kevin Leclerc
- NYU Grossman School of Medicine, NYU Langone Orthopedics, New York, NY 10016, USA
| | - Lindsey H. Remark
- NYU Grossman School of Medicine, NYU Langone Orthopedics, New York, NY 10016, USA
| | - Emma Muiños Lopeź
- NYU Grossman School of Medicine, NYU Langone Orthopedics, New York, NY 10016, USA
| | - Philipp Leucht
- NYU Grossman School of Medicine, NYU Langone Orthopedics, New York, NY 10016, USA
- NYU Grossman School of Medicine, Department of Cell Biology, New York, NY 10016, USA
| |
Collapse
|
8
|
Yahara Y, Ma X, Gracia L, Alman BA. Monocyte/Macrophage Lineage Cells From Fetal Erythromyeloid Progenitors Orchestrate Bone Remodeling and Repair. Front Cell Dev Biol 2021; 9:622035. [PMID: 33614650 PMCID: PMC7889961 DOI: 10.3389/fcell.2021.622035] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/12/2021] [Indexed: 12/21/2022] Open
Abstract
A third of the population sustains a bone fracture, and the pace of fracture healing slows with age. The slower pace of repair is responsible for the increased morbidity in older individuals who sustain a fracture. Bone healing progresses through overlapping phases, initiated by cells of the monocyte/macrophage lineage. The repair process ends with remodeling. This last phase is controlled by osteoclasts, which are bone-specific multinucleated cells also of the monocyte/macrophage lineage. The slower rate of healing in aging can be rejuvenated by macrophages from young animals, and secreted proteins from macrophage regulate undifferentiated mesenchymal cells to become bone-forming osteoblasts. Macrophages can derive from fetal erythromyeloid progenitors or from adult hematopoietic progenitors. Recent studies show that fetal erythromyeloid progenitors are responsible for the osteoclasts that form the space in bone for hematopoiesis and the fetal osteoclast precursors reside in the spleen postnatally, traveling through the blood to participate in fracture repair. Differences in secreted proteins between macrophages from old and young animals regulate the efficiency of osteoblast differentiation from undifferentiated mesenchymal precursor cells. Interestingly, during the remodeling phase osteoclasts can form from the fusion between monocyte/macrophage lineage cells from the fetal and postnatal precursor populations. Data from single cell RNA sequencing identifies specific markers for populations derived from the different precursor populations, a finding that can be used in future studies. Here, we review the diversity of macrophages and osteoclasts, and discuss recent finding about their developmental origin and functions, which provides novel insights into their roles in bone homeostasis and repair.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, Japan.,Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Xinyi Ma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Liam Gracia
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
9
|
Moore ER, Mathews OA, Yao Y, Yang Y. Prx1-expressing cells contributing to fracture repair require primary cilia for complete healing in mice. Bone 2021; 143:115738. [PMID: 33188955 PMCID: PMC7769995 DOI: 10.1016/j.bone.2020.115738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/25/2020] [Accepted: 11/07/2020] [Indexed: 02/09/2023]
Abstract
Bone is a dynamic organ that is continuously modified during development, load-induced adaptation, and fracture repair. Understanding the cellular and molecular mechanisms for natural fracture healing can lead to therapeutics that enhance the quality of newly formed tissue, advance the rate of healing, or replace the need for invasive surgical procedures. Prx1-expressing cells in the periosteum are thought to supply the majority of osteoblasts and chondrocytes in the fracture callus, but the exact mechanisms for this behavior are unknown. The primary cilium is a sensory organelle that is known to mediate several signaling pathways involved in fracture healing and required for Prx1-expressing cells to contribute to juvenile bone development and adult load-induced bone formation. We therefore investigated the role of Prx1-expressing cell primary cilia in fracture repair by developing a mouse model that enabled us to simultaneously track Prx1 lineage cell fate and disrupt Prx1-expressing cell primary cilia in vivo. The cilium KO mice exhibited abnormally large calluses with significantly decreased bone formation and persistent cartilage nodules. Analysis of mRNA expression in the early soft callus revealed downregulation of osteogenesis, Hh signaling, and Wnt signaling, and upregulation of chondrogenesis and angiogenesis. The mutant mice also exhibited decreased Osx and Periostin but increased αSMA and PECAM-1 protein expression in the hard callus. We further used a Gli1LacZ reporter and found that Hh signaling was significantly upregulated in the mutant callus at later stages of healing. Interestingly, altered protein expression and Hh signaling did not correlate with labeled Prx1-lineage cells, suggesting loss of cilia altered Hh signaling non-autonomously. Overall, cilium KO mice demonstrated severely delayed and incomplete fracture healing, and our findings suggest Prx1-expressing cell primary cilia are necessary to tune Hh signaling for proper fracture repair.
Collapse
Affiliation(s)
| | - O Amandhi Mathews
- Harvard School of Dental Medicine, Boston, MA, USA; University of Dallas, Irving, TX, USA
| | - Yichen Yao
- Harvard School of Dental Medicine, Boston, MA, USA; Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingzi Yang
- Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
10
|
Abstract
The most common procedure that has been developed for use in rats and mice to model fracture healing is described. The nature of the regenerative processes that may be assessed and the types of research questions that may be addressed with this model are briefly outlined. The detailed surgical protocol to generate closed simple transverse fractures is presented and general considerations when setting up an experiment using this model are described.
Collapse
|
11
|
Influence of the TGF-β Superfamily on Osteoclasts/Osteoblasts Balance in Physiological and Pathological Bone Conditions. Int J Mol Sci 2020; 21:ijms21207597. [PMID: 33066607 PMCID: PMC7589189 DOI: 10.3390/ijms21207597] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/19/2022] Open
Abstract
The balance between bone forming cells (osteoblasts/osteocytes) and bone resorbing cells (osteoclasts) plays a crucial role in tissue homeostasis and bone repair. Several hormones, cytokines, and growth factors-in particular the members of the TGF-β superfamily such as the bone morphogenetic proteins-not only regulate the proliferation, differentiation, and functioning of these cells, but also coordinate the communication between them to ensure an appropriate response. Therefore, this review focuses on TGF-β superfamily and its influence on bone formation and repair, through the regulation of osteoclastogenesis, osteogenic differentiation of stem cells, and osteoblasts/osteoclasts balance. After introducing the main types of bone cells, their differentiation and cooperation during bone remodeling and fracture healing processes are discussed. Then, the TGF-β superfamily, its signaling via canonical and non-canonical pathways, as well as its regulation by Wnt/Notch or microRNAs are described and discussed. Its important role in bone homeostasis, repair, or disease is also highlighted. Finally, the clinical therapeutic uses of members of the TGF-β superfamily and their associated complications are debated.
Collapse
|
12
|
Abstract
The skeleton is highly vascularized due to the various roles blood vessels play in the homeostasis of bone and marrow. For example, blood vessels provide nutrients, remove metabolic by-products, deliver systemic hormones, and circulate precursor cells to bone and marrow. In addition to these roles, bone blood vessels participate in a variety of other functions. This article provides an overview of the afferent, exchange and efferent vessels in bone and marrow and presents the morphological layout of these blood vessels regarding blood flow dynamics. In addition, this article discusses how bone blood vessels participate in bone development, maintenance, and repair. Further, mechanical loading-induced bone adaptation is presented regarding interstitial fluid flow and pressure, as regulated by the vascular system. The role of the sympathetic nervous system is discussed in relation to blood vessels and bone. Finally, vascular participation in bone accrual with intermittent parathyroid hormone administration, a medication prescribed to combat age-related bone loss, is described and age- and disease-related impairments in blood vessels are discussed in relation to bone and marrow dysfunction. © 2020 American Physiological Society. Compr Physiol 10:1009-1046, 2020.
Collapse
Affiliation(s)
- Rhonda D Prisby
- Bone Vascular and Microcirculation Laboratory, Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
13
|
Clark D, Brazina S, Yang F, Hu D, Hsieh CL, Niemi EC, Miclau T, Nakamura MC, Marcucio R. Age-related changes to macrophages are detrimental to fracture healing in mice. Aging Cell 2020; 19:e13112. [PMID: 32096907 PMCID: PMC7059136 DOI: 10.1111/acel.13112] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/05/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
The elderly population suffers from higher rates of complications during fracture healing that result in increased morbidity and mortality. Inflammatory dysregulation is associated with increased age and is a contributing factor to the myriad of age-related diseases. Therefore, we investigated age-related changes to an important cellular regulator of inflammation, the macrophage, and the impact on fracture healing outcomes. We demonstrated that old mice (24 months) have delayed fracture healing with significantly less bone and more cartilage compared to young mice (3 months). The quantity of infiltrating macrophages into the fracture callus was similar in old and young mice. However, RNA-seq analysis demonstrated distinct differences in the transcriptomes of macrophages derived from the fracture callus of old and young mice, with an up-regulation of M1/pro-inflammatory genes in macrophages from old mice as well as dysregulation of other immune-related genes. Preventing infiltration of the fracture site by macrophages in old mice improved healing outcomes, with significantly more bone in the calluses of treated mice compared to age-matched controls. After preventing infiltration by macrophages, the macrophages remaining within the fracture callus were collected and examined via RNA-seq analysis, and their transcriptome resembled macrophages from young calluses. Taken together, infiltrating macrophages from old mice demonstrate detrimental age-related changes, and depleting infiltrating macrophages can improve fracture healing in old mice.
Collapse
Affiliation(s)
- Daniel Clark
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
- Division of PeriodontologyDepartment of Orofacial SciencesSchool of DentistryUniversity of California San FranciscoSan FranciscoCAUSA
| | - Sloane Brazina
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Frank Yang
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Diane Hu
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Christine L. Hsieh
- Division of RheumatologyDepartment of MedicineSan Francisco VA Health Care SystemSan FranciscoCAUSA
| | - Erene C. Niemi
- Division of RheumatologyDepartment of MedicineSan Francisco VA Health Care SystemSan FranciscoCAUSA
| | - Theodore Miclau
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Mary C. Nakamura
- Division of RheumatologyDepartment of MedicineSan Francisco VA Health Care SystemSan FranciscoCAUSA
| | - Ralph Marcucio
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| |
Collapse
|
14
|
Gulbrandsen TR, Hulick RM, Polk AJ, Weldy JM, Howell KL, Spitler CA, Crist BD. Does surgical approach affect sagittal plane alignment and pilon fracture outcomes? Injury 2020; 51:750-758. [PMID: 32008815 DOI: 10.1016/j.injury.2020.01.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/07/2020] [Accepted: 01/19/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE Sagittal plane alignment beyond articular reduction and its effect on clinical outcomes has not been emphasized. Surgical approach may influence a surgeon's ability to correct the sagittal plane alignment. The purpose of our study was to evaluate how surgical approach impacts anterior distal tibial angle (ADTA) and lateral talar station (LTS). Our hypothesis was that the anterolateral (AL) approach would improve the sagittal plane parameters due to the primary plate placement. PATIENTS AND METHODS A retrospective review was performed on patients who underwent operative management for pilon fractures at 2 ACS Level 1 Academic Trauma Centers. Clinical data points including demographics, comorbidities, AO/OTA classification, surgical approach, and complications were recorded. Quality of reduction was measured using the ADTA, lateral distal tibia angle (LDTA), and lateral talar station (LTS) from radiographs. RESULTS 580 pilon fractures met inclusion criteria. When compared to the AL approach, the modified anteromedial (AM) approach had decreased rates of local wound care, and unplanned reoperations. The AM approach had increased rates of superficial infection, deep infection, non-union, and amputations. There was no difference in ADTA, LDTA, or LTS between the AM and AL approach (P = 0.49, P = 0.41, P = 0.85). There was a difference in LTS with tobacco users (P = 0.02). CONCLUSIONS The sagittal plane alignment does not appear to be affected by the surgical approach. Therefore, the surgical approach to pilon fractures should be based on the fracture pattern and the patient's soft tissue envelope. This study shows that the AM is a relatively safe and effective approach to complex fractures and the surgeon should consider the specific fracture pattern and patient soft tissue envelope when choosing the specific approach.
Collapse
Affiliation(s)
- Trevor R Gulbrandsen
- University of Iowa Hospitals and Clinics, Department of Orthopedic Surgery, Iowa City, IA, United States
| | - Robert M Hulick
- University of Mississippi Medical Center, Department of Orthopaedic Surgery, Jackson, MS, United States
| | - Andrew J Polk
- University of Missouri School of Medicine, Columbia, MO, United States
| | - John M Weldy
- University of Mississippi Medical Center, Department of Orthopaedic Surgery, Jackson, MS, United States
| | - Kathryn L Howell
- Tulane University, Department of Orthopaedic Surgery, New Orleans, LA, United States
| | - Clay A Spitler
- University of Mississippi Medical Center, Department of Internal Medicine, Jackson, MS, United States; University of Alabama-Birmingham, Department of Orthopaedic Surgery, Birmingham, AL, United States
| | - Brett D Crist
- University of Missouri, Department of Orthopaedic Surgery, Columbia, MO, United States.
| |
Collapse
|
15
|
Ras associated with diabetes may play a role in fracture nonunion development in rats. BMC Musculoskelet Disord 2019; 20:602. [PMID: 31830958 PMCID: PMC6909478 DOI: 10.1186/s12891-019-2970-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Rad is the prototypic member of a subfamily of Ras-related small G-proteins and is highly expressed in the skeletal muscle of patients with type II diabetes. Our previous microarray analysis suggested that Rad may mediate fracture nonunion development. Thus, the present study used rat experimental models to investigate and compare the gene and protein expression patterns of both Rad and Rem1, another RGK subfamily member, in nonunions and standard healing fractures. Methods Standard healing fractures and nonunions (produced via periosteal cauterization at the fracture site) were created in the femurs of 3-month-old male Sprague-Dawley rats. At post-fracture days 7, 14, 21, and 28, the fracture callus and fibrous tissue from the standard healing fractures and nonunions, respectively, were harvested and screened (via real-time PCR) for Rad and Rem1 expression. The immunolocalization of both encoded proteins was analyzed at post-fracture days 14 and 21. At the same time points, hematoxylin and eosin staining was performed to identify the detailed tissue structures. Results Results of real-time PCR analysis showed that Rad expression increased significantly in the nonunions, compared to that in the standard healing fractures, at post-fracture days 14, 21, and 28. Conversely, immunohistochemical analysis revealed the immunolocalization of Rad to be similar to that of Rem1 in both fracture types at post-fracture days 14 and 21. Conclusions Rad may mediate nonunion development, and thus, may be a promising therapeutic target to treat these injuries.
Collapse
|
16
|
Kim M, Kang JH, Oh GH, Park MH. Ishige sinicola extract stimulates osteoblast proliferation and differentiation via the bone morphogenetic protein 2/runt-related gene 2 signalling pathway. Z NATURFORSCH C 2019; 74:167-174. [PMID: 31085751 DOI: 10.1515/znc-2018-0044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/16/2019] [Indexed: 12/17/2023]
Abstract
Osteoporosis is one of the most common bone diseases, occurring due to an imbalance between bone formation and bone resorption. The aim of this study was to investigate the effects of Ishige sinicola, a brown alga, on osteoblast differentiation through the activation of the bone morphogenetic protein 2 (BMP-2)/runt-related transcription factor 2 (Runx2) signalling pathway in MC3T3-E1 cells. A cell proliferation assay, alkaline phosphatase (ALP) activity assay, alizarin red staining, and expression analysis of osteoblastic genes were carried out to assess MC3T3-E1 cell proliferation and osteoblastic differentiation. We found that I. sinicola extract (ISE) increased cell proliferation in a dose-dependent manner. Ishige sinicola extract markedly promoted ALP activity and mineralization. Alizarin red S staining demonstrated that ISE treatment tended to increase extracellular matrix calcium accumulation. Moreover, ISE up-regulated the osteoprotegerin/receptor activator of nuclear factor κB ligand ratio. Ishige sinicola extract also increased the protein expression levels of type 1 collagen, ALP, osteocalcin, osterix, BMP-2, and Runx2. Therefore, ISE showed potential in stimulating osteoblastic bone formation, and it might be useful for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Mihyang Kim
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 46958, Republic of Korea
| | - Jeong Hyeon Kang
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 46958, Republic of Korea
| | - Geun Hye Oh
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 46958, Republic of Korea
| | - Mi Hwa Park
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 46958, Republic of Korea, Phone: +82-51-999-5908, Fax: +82-51-999-5457
| |
Collapse
|
17
|
Wagner DR, Karnik S, Gunderson ZJ, Nielsen JJ, Fennimore A, Promer HJ, Lowery JW, Loghmani MT, Low PS, McKinley TO, Kacena MA, Clauss M, Li J. Dysfunctional stem and progenitor cells impair fracture healing with age. World J Stem Cells 2019; 11:281-296. [PMID: 31293713 PMCID: PMC6600851 DOI: 10.4252/wjsc.v11.i6.281] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/26/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Successful fracture healing requires the simultaneous regeneration of both the bone and vasculature; mesenchymal stem cells (MSCs) are directed to replace the bone tissue, while endothelial progenitor cells (EPCs) form the new vasculature that supplies blood to the fracture site. In the elderly, the healing process is slowed, partly due to decreased regenerative function of these stem and progenitor cells. MSCs from older individuals are impaired with regard to cell number, proliferative capacity, ability to migrate, and osteochondrogenic differentiation potential. The proliferation, migration and function of EPCs are also compromised with advanced age. Although the reasons for cellular dysfunction with age are complex and multidimensional, reduced expression of growth factors, accumulation of oxidative damage from reactive oxygen species, and altered signaling of the Sirtuin-1 pathway are contributing factors to aging at the cellular level of both MSCs and EPCs. Because of these geriatric-specific issues, effective treatment for fracture repair may require new therapeutic techniques to restore cellular function. Some suggested directions for potential treatments include cellular therapies, pharmacological agents, treatments targeting age-related molecular mechanisms, and physical therapeutics. Advanced age is the primary risk factor for a fracture, due to the low bone mass and inferior bone quality associated with aging; a better understanding of the dysfunctional behavior of the aging cell will provide a foundation for new treatments to decrease healing time and reduce the development of complications during the extended recovery from fracture healing in the elderly.
Collapse
Affiliation(s)
- Diane R Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Zachary J Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States
| | - Alanna Fennimore
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Hunter J Promer
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - M Terry Loghmani
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, United States
| | - Matthias Clauss
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jiliang Li
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| |
Collapse
|
18
|
Meinberg EG, Clark D, Miclau KR, Marcucio R, Miclau T. Fracture repair in the elderly: Clinical and experimental considerations. Injury 2019; 50 Suppl 1:S62-S65. [PMID: 31130210 PMCID: PMC7021229 DOI: 10.1016/j.injury.2019.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/01/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023]
Abstract
Fractures in the elderly represent a significant and rising socioeconomic problem. Although aging has been associated with delays in healing, there is little direct clinical data isolating the effects of aging on bone healing from the associated comorbidities that are frequently present in elderly populations. Basic research has demonstrated that all of the components of fracture repair-cells, extracellular matrix, blood supply, and molecules and their receptors-are negatively impacted by the aging process, which likely explains poorer clinical outcomes. Improved understanding of age-related fracture healing should aid in the development of novel treatment strategies, technologies, and therapies to improve bone repair in elderly patients.
Collapse
Affiliation(s)
- E G Meinberg
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - D Clark
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - K R Miclau
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - R Marcucio
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - T Miclau
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA.
| |
Collapse
|
19
|
|
20
|
McKenzie JA, Maschhoff C, Liu X, Migotsky N, Silva MJ, Gardner MJ. Activation of hedgehog signaling by systemic agonist improves fracture healing in aged mice. J Orthop Res 2019; 37:51-59. [PMID: 29663560 PMCID: PMC6226344 DOI: 10.1002/jor.24017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/04/2018] [Indexed: 02/04/2023]
Abstract
Fracture healing is a complex process of many coordinated biological pathways. This system can go awry resulting in nonunion, which leads to significant patient morbidity. The Hedgehog (Hh) signaling pathway is upregulated in fracture healing. We hypothesized that the Hh signaling pathway can be pharmacologically modulated to positively affect fracture healing. Diaphyseal femur fractures were created in elderly mice (18 months, C57BL/6 females), which have a blunted and delayed healing response compared to younger mice, and were stabilized with intramedullary pins. To activate the Hh pathway we targeted the receptor Smoothened using an agonist (Hh-Ag1.5 [Hh-Ag]) and compared this to a vehicle control. Expression of Hh target genes were significantly increased in the fracture callus of the agonist group compared to controls, indicating pathway activation. Expression of osteogenic and chondrogenic-related genes was greatly upregulated in fracture callus versus intact femora, although Hh agonist treatment did not consistently enhance this response. Blindly graded, radiographic callus healing scores were significantly higher in the Hh-Ag groups at post operative day (POD) 14, indicating earlier callus bridging. On microCT, Hh-Ag treatment led to greater callus volume (+40%) and bone volume (+25%) at POD21. By day 14, callus vascularity, as assessed by 3D microCT angiography vessel volume, was 85% greater in the Hh-Ag group. Finally, mechanical strength of the calluses in the Hh-Ag groups was significantly greater than in the control groups at POD21. In conclusion, systemic administration of a Hh agonist appears to improve the osseous and vascular healing responses in a mouse fracture healing-impaired model. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
| | | | - Xiaochen Liu
- Washington University Orthopedics, St. Louis, MO
| | | | | | - Michael J. Gardner
- Washington University Orthopedics, St. Louis, MO,Stanford University Orthopedics, Stanford, CA
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW This review summarizes research on the physiological changes that occur with aging and the resulting effects on fracture healing. RECENT FINDINGS Aging affects the inflammatory response during fracture healing through senescence of the immune response and increased systemic pro-inflammatory status. Important cells of the inflammatory response, macrophages, T cells, mesenchymal stem cells, have demonstrated intrinsic age-related changes that could impact fracture healing. Additionally, vascularization and angiogenesis are impaired in fracture healing of the elderly. Finally, osteochondral cells and their progenitors demonstrate decreased activity and quantity within the callus. Age-related changes affect many of the biologic processes involved in fracture healing. However, the contributions of such changes do not fully explain the poorer healing outcomes and increased morbidity reported in elderly patients. Future research should address this gap in understanding in order to provide improved and more directed treatment options for the elderly population.
Collapse
Affiliation(s)
- Dan Clark
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California at San Francisco, 513 Parnassus Ave., Rm. S-619A, San Francisco, CA, 94143, USA
| | - Mary Nakamura
- Department of Medicine, University of California at San Francisco, San Francisco, CA, 94143-0451, USA
- Department of Pathology, VA Medical Center, University of California San Francisco & Pathology Service, San Francisco, CA, 94121, USA
| | - Ted Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA.
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA.
| |
Collapse
|
22
|
Komrakova M, Hoffmann DB, Nuehnen V, Stueber H, Wassmann M, Wicke M, Tezval M, Stuermer KM, Sehmisch S. The Effect of Vibration Treatments Combined with Teriparatide or Strontium Ranelate on Bone Healing and Muscle in Ovariectomized Rats. Calcif Tissue Int 2016; 99:408-22. [PMID: 27272029 DOI: 10.1007/s00223-016-0156-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/23/2016] [Indexed: 01/14/2023]
Abstract
The aim of the present study was to study the effect of combined therapy of teriparatide (PTH) or strontium ranelate (SR) with whole-body vibration (WBV) on bone healing and muscle properties in an osteopenic rat model. Seventy-two rats (3 months old) were bilaterally ovariectomized (Ovx), and 12 rats were left intact (Non-Ovx). After 8 weeks, bilateral transverse osteotomy was performed at the tibia metaphysis in all rats. Thereafter, Ovx rats were divided into six groups (n = 12): (1) Ovx-no treatment, (2) Ovx + vibration (Vib), (3) SR, (4) SR + Vib, (5) PTH, and (6) PTH + Vib. PTH (40 μg/kg BW sc. 5×/week) and SR (613 mg/kg BW in food daily) were applied on the day of ovariectomy, vibration treatments 5 days later (vertical, 70 Hz, 0.5 mm, 2×/day for 15 min) for up to 6 weeks. In the WBV + SR group, the callus density, trabecular number, and Alp and Oc gene expression were decreased compared to SR alone. In the WBV + PTH group, the cortical and callus widths, biomechanical properties, Opg gene expression, and Opg/Rankl ratio were increased; the cortical and callus densities were decreased compared to PTH alone. A case of non-bridging was found in both vibrated groups. Vibration alone did not change the bone parameters; PTH possessed a stronger effect than SR therapy. In muscles, combined therapies improved the fiber size of Ovx rats. WBV could be applied alone or in combination with anti-osteoporosis drug therapy to improve muscle tissue. However, in patients with fractures, anti-osteoporosis treatments and the application of vibration could have an adverse effect on bone healing.
Collapse
Affiliation(s)
- M Komrakova
- Department of Trauma Surgery and Reconstructive Surgery, University Medicine of Goettingen, Robert-Koch Str. 40, 37075, Göttingen, Germany.
| | - D B Hoffmann
- Department of Trauma Surgery and Reconstructive Surgery, University Medicine of Goettingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - V Nuehnen
- Department of Trauma Surgery and Reconstructive Surgery, University Medicine of Goettingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - H Stueber
- Department of Trauma Surgery and Reconstructive Surgery, University Medicine of Goettingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - M Wassmann
- Department of Medical Microbiology, Subdivision of General Hygiene and Environmental Health, University of Goettingen, Humboldallee 34a, 37073, Göttingen, Germany
| | - M Wicke
- Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - M Tezval
- Department of Trauma Surgery and Reconstructive Surgery, University Medicine of Goettingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - K M Stuermer
- Department of Trauma Surgery and Reconstructive Surgery, University Medicine of Goettingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - S Sehmisch
- Department of Trauma Surgery and Reconstructive Surgery, University Medicine of Goettingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| |
Collapse
|
23
|
Histing T, Heerschop K, Klein M, Scheuer C, Stenger D, Herath SC, Pohlemann T, Menger MD. Effect of Stabilization on the Healing Process of Femur Fractures in Aged Mice. J INVEST SURG 2016; 29:202-8. [PMID: 26891453 DOI: 10.3109/08941939.2015.1127448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The influence of mechanical stability on fracture healing has previously been studied in adult mice, but is poorly understood in aged animals. Therefore, we herein studied the effect of stabilization on the healing process of femur fractures in aged mice. METHODS Twenty-four 18-month-old CD-1 mice were stabilized after midshaft fracture of the femur with an intramedullary screw. In another 24 18-month-old mice, the femur fractures were left unstabilized. Bone healing was studied by radiological, biomechanical, histomorphometric, and protein expression analyses. RESULTS After 2 and 5 weeks of healing, the callus of nonstabilized fractures compared to stabilized fractures was significantly larger, containing a significantly smaller amount of osseous tissue and a higher amount of cartilaginous tissue. This was associated with a significantly lower biomechanical stiffness during the early phase of healing. However, during the late phase of fracture healing both nonstabilized and stabilized fractures showed a biomechanical stiffness of ∼40%. Of interest, Western blot analyses of callus tissue demonstrated that the expression of proteins related to angiogenesis, bone formation and remodeling, i.e. VEGF, CYR61, BMP-2, BMP-4, Col-2, Col-10, RANKL, OPG, did not differ between nonstabilized and stabilized fractures. CONCLUSION Nonstabilized fractures in aged mice show delayed healing and remodeling. This is not caused by an altered protein expression in the callus but rather by the excessive interfragmentary movements.
Collapse
Affiliation(s)
- T Histing
- a Department of Trauma, Hand and Reconstructive Surgery , Saarland University , D-66421 Homburg/Saar, Germany
| | - K Heerschop
- a Department of Trauma, Hand and Reconstructive Surgery , Saarland University , D-66421 Homburg/Saar, Germany
| | - M Klein
- a Department of Trauma, Hand and Reconstructive Surgery , Saarland University , D-66421 Homburg/Saar, Germany
| | - C Scheuer
- b Institute for Clinical & Experimental Surgery , Saarland University , Homburg/Saar, Germany
| | - D Stenger
- a Department of Trauma, Hand and Reconstructive Surgery , Saarland University , D-66421 Homburg/Saar, Germany
| | - S C Herath
- a Department of Trauma, Hand and Reconstructive Surgery , Saarland University , D-66421 Homburg/Saar, Germany
| | - T Pohlemann
- a Department of Trauma, Hand and Reconstructive Surgery , Saarland University , D-66421 Homburg/Saar, Germany
| | - M D Menger
- b Institute for Clinical & Experimental Surgery , Saarland University , Homburg/Saar, Germany
| |
Collapse
|
24
|
Waki T, Lee SY, Niikura T, Iwakura T, Dogaki Y, Okumachi E, Kuroda R, Kurosaka M. Profiling microRNA expression in fracture nonunions: Potential role of microRNAs in nonunion formation studied in a rat model. Bone Joint J 2015. [PMID: 26224835 DOI: 10.1302/0301-620x.97b8.34966] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
MicroRNAs (miRNAs ) are small non-coding RNAs that regulate gene expression. We hypothesised that the functions of certain miRNAs and changes to their patterns of expression may be crucial in the pathogenesis of nonunion. Healing fractures and atrophic nonunions produced by periosteal cauterisation were created in the femora of 94 rats, with 1:1 group allocation. At post-fracture days three, seven, ten, 14, 21 and 28, miRNAs were extracted from the newly generated tissue at the fracture site. Microarray and real-time polymerase chain reaction (PCR) analyses of day 14 samples revealed that five miRNAs, miR-31a-3p, miR-31a-5p, miR-146a-5p, miR-146b-5p and miR-223-3p, were highly upregulated in nonunion. Real-time PCR analysis further revealed that, in nonunion, the expression levels of all five of these miRNAs peaked on day 14 and declined thereafter. Our results suggest that miR-31a-3p, miR-31a-5p, miR-146a-5p, miR-146b-5p and miR-223-3p may play an important role in the development of nonunion. These findings add to the understanding of the molecular mechanism for nonunion formation and may lead to the development of novel therapeutic strategies for its treatment.
Collapse
Affiliation(s)
- T Waki
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - S Y Lee
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - T Niikura
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - T Iwakura
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Y Dogaki
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - E Okumachi
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - R Kuroda
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - M Kurosaka
- Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
25
|
Komrakova M, Weidemann A, Dullin C, Ebert J, Tezval M, Stuermer KM, Sehmisch S. The Impact of Strontium Ranelate on Metaphyseal Bone Healing in Ovariectomized Rats. Calcif Tissue Int 2015; 97:391-401. [PMID: 26084691 DOI: 10.1007/s00223-015-0019-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/01/2015] [Indexed: 11/24/2022]
Abstract
The following questions were addressed: whether therapy with strontium ranelate (SR) should be continued or interrupted if the fractures occur during SR treatment and whether SR could be applied directly after fracture to improve bone healing. Sprague-Dawley rats (3 month old) were ovariectomized (Ovx, n = 48) or left intact (n = 12). After 8 weeks, a bilateral transverse osteotomy of the tibia metaphysis was created in all rats. Ovx rats were divided into four groups: Ovx; SR applied directly after Ovx until osteotomy (prophylaxis, SR pr, 8 weeks); SR applied after osteotomy (therapy, SR th, 5 weeks); SR applied during the whole experiment (pr + th, 13 weeks). SR dosage was 625 mg/kg body weight/day, administered in the feed. Five weeks later, tibiae were analyzed by biomechanical, histological, micro-CT, and gene expression analyses. The SR pr + th treatment increased total bone mineral density (BMD), bone volume fraction, cortical BMD and volume, callus area and density, serum alkaline phosphatase, tartrate-resistant acid phosphatase mRNA, accelerated osteotomy bridging, and callus formation at weeks 2 and 3 of healing and decreased the osteoprotegerin/receptor activator of nuclear factor kB ligand mRNA ratio. SR th enlarged callus area and improved callus formation during the 5th week of healing. SR pr improved cortical BMD preserving bone after SR discontinuation (5-week rest); the bone healing was not affected. SR content in the tibia metaphysis was the highest in SR pr + th group and was not different between SR pr and SR th. SR has a positive effect on osteoporotic bone healing in rat and SR treatment can be continued after the fracture occurs or applied directly after the fracture.
Collapse
Affiliation(s)
- Marina Komrakova
- Department of Trauma Surgery and Reconstructive Surgery, University Medical Center Göttingen, Robert-Koch St. 40, 37075, Göttingen, Germany.
| | - Anna Weidemann
- Department of Trauma Surgery and Reconstructive Surgery, University Medical Center Göttingen, Robert-Koch St. 40, 37075, Göttingen, Germany
| | - Christian Dullin
- Department of Radiology, University of Göttingen, Robert-Koch 40, 37075, Göttingen, Germany
| | - Joachim Ebert
- Department of Medical Microbiology, Subdivision of General Hygiene and Environmental Health, University of Göttingen, Humboldallee 34a, 37073, Göttingen, Germany
| | - Mohammad Tezval
- Department of Trauma Surgery and Reconstructive Surgery, University Medical Center Göttingen, Robert-Koch St. 40, 37075, Göttingen, Germany
| | - Klaus Michael Stuermer
- Department of Trauma Surgery and Reconstructive Surgery, University Medical Center Göttingen, Robert-Koch St. 40, 37075, Göttingen, Germany
| | - Stephan Sehmisch
- Department of Trauma Surgery and Reconstructive Surgery, University Medical Center Göttingen, Robert-Koch St. 40, 37075, Göttingen, Germany
| |
Collapse
|
26
|
Xia W, Zhang F, Xie C, Jiang M, Hou M. Macrophage migration inhibitory factor confers resistance to senescence through CD74-dependent AMPK-FOXO3a signaling in mesenchymal stem cells. Stem Cell Res Ther 2015; 6:82. [PMID: 25896286 PMCID: PMC4453287 DOI: 10.1186/s13287-015-0076-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 12/15/2014] [Accepted: 04/10/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs)-based therapies have had positive outcomes in animal models of cardiovascular diseases. However, the number and function of MSCs decline with age, reducing their ability to contribute to endogenous injury repair. The potential of stem cells to restore damaged tissue in older individuals can be improved by specific pretreatment aimed at delaying senescence and improving their regenerative properties. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that modulates age-related signaling pathways, and hence is a good candidate for rejuvenative function. Methods Bone marrow-derived mesenchymal stem cells (BM-MSCs) were isolated from young (6-month-old) or aged (24-month-old) male donor rats. Cell proliferation was measured using the CCK8 cell proliferation assay; secretion of VEGF, bFGF, HGF, and IGF was assessed by RT-qPCR and ELISA. Apoptosis was induced by hypoxia and serum deprivation (hypoxia/SD) for up to 6 hr, and examined by flow cytometry. Expression levels of AMP-activated protein kinase (AMPK) and forkhead box class O 3a (FOXO3a) were detected by Western blotting. CD74 expression was assayed using RT-qPCR, Western blotting, and immunofluorescence. Results In this study, we found that MSCs isolated from the bone marrow of aged rats displayed reduced proliferative capacity, impaired ability to mediate paracrine signaling, and lower resistance to hypoxia/serum deprivation-induced apoptosis, when compared to younger MSCs. Interestingly, pretreatment of aged MSCs with MIF enhanced their growth, paracrine function and survival. We detected enhanced secretion of VEGF, bFGF, HGF, and IGF from MIF-treated MSCs using ELISA. Finally, we show that hypoxia/serum deprivation-induced apoptosis is inhibited in aged MSCs following MIF exposure. Next, we found that the mechanism underlying the rejuvenating function of MIF involves increased CD74-dependent phosphorylation of AMPK and FOXO3a. Furthermore, this effect was abolished when CD74, AMPK, or FOXO3a expression was silenced using small-interfering RNAs(siRNA). Conclusions MIF can rejuvenate MSCs from a state of age-induced senescence by interacting with CD74 and subsequently activating AMPK-FOXO3a signaling pathways. Pretreatment of MSCs with MIF may have important therapeutic implications in restoration or rejuvenation of endogenous bone marrow-MSCs in aged individuals.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Fengyun Zhang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150086, PR China. .,Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150086, PR China.
| | - Congying Xie
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Miaomiao Jiang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150086, PR China. .,Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150086, PR China.
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, PR China.
| |
Collapse
|
27
|
Transforming growth factor Beta family: insight into the role of growth factors in regulation of fracture healing biology and potential clinical applications. Mediators Inflamm 2015; 2015:137823. [PMID: 25709154 PMCID: PMC4325469 DOI: 10.1155/2015/137823] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/09/2014] [Indexed: 01/15/2023] Open
Abstract
The transforming growth factor beta (TGF-β) family forms a group of three isoforms, TGF-β1, TGF-β2, and TGF-β3, with their structure formed by interrelated dimeric polypeptide chains. Pleiotropic and redundant functions of the TGF-β family concern control of numerous aspects and effects of cell functions, including proliferation, differentiation, and migration, in all tissues of the human body. Amongst many cytokines and growth factors, the TGF-β family is considered a group playing one of numerous key roles in control of physiological phenomena concerning maintenance of metabolic homeostasis in the bone tissue. By breaking the continuity of bone tissue, a spread-over-time and complex bone healing process is initiated, considered a recapitulation of embryonic intracartilaginous ossification. This process is a cascade of local and systemic phenomena spread over time, involving whole cell lineages and various cytokines and growth factors. Numerous in vivo and in vitro studies in various models analysing cytokines and growth factors' involvement have shown that TGF-β has a leading role in the fracture healing process. This paper sums up current knowledge on the basis of available literature concerning the role of the TGF-β family in the fracture healing process.
Collapse
|
28
|
Lopas LA, Belkin NS, Mutyaba PL, Gray CF, Hankenson KD, Ahn J. Fractures in geriatric mice show decreased callus expansion and bone volume. Clin Orthop Relat Res 2014; 472:3523-32. [PMID: 25106797 PMCID: PMC4182401 DOI: 10.1007/s11999-014-3829-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 07/15/2014] [Indexed: 01/31/2023]
Abstract
BACKGROUND Poor fracture healing in geriatric populations is a significant source of morbidity, mortality, and cost to individuals and society; however, a fundamental biologic understanding of age-dependent healing remains elusive. The development of an aged-based fracture model system would allow for a mechanistic understanding that could guide future biologic treatments. QUESTIONS/PURPOSES Using a small animal model of long-bone fracture healing based on chronologic age, we asked how aging affected (1) the amount, density, and proportion of bone formed during healing; (2) the amount of cartilage produced and the progression to bone during healing; (3) the callus structure and timing of the fracture healing; and (4) the behavior of progenitor cells relative to the observed deficiencies of geriatric fracture healing. METHODS Transverse, traumatic tibial diaphyseal fractures were created in 5-month-old (n=104; young adult) and 25-month-old (n=107; which we defined as geriatric, and are approximately equivalent to 70-85 year-old humans) C57BL/6 mice. Fracture calluses were harvested at seven times from 0 to 40 days postfracture for micro-CT analysis (total volume, bone volume, bone volume fraction, connectivity density, structure model index, trabecular number, trabecular thickness, trabecular spacing, total mineral content, bone mineral content, tissue mineral density, bone mineral density, degree of anisotropy, and polar moment of inertia), histomorphometry (total callus area, cartilage area, percent of cartilage, hypertrophic cartilage area, percent of hypertrophic cartilage area, bone and osteoid area, percent of bone and osteoid area), and gene expression quantification (fold change). RESULTS The geriatric mice produced a less robust healing response characterized by a pronounced decrease in callus amount (mean total volume at 20 days postfracture, 30.08±11.53 mm3 versus 43.19±18.39 mm3; p=0.009), density (mean bone mineral density at 20 days postfracture, 171.14±64.20 mg hydroxyapatite [HA]/cm3 versus 210.79±37.60 mg HA/cm3; p=0.016), and less total cartilage (mean cartilage area at 10 days postfracture, 101,279±46,755 square pixels versus 302,167±137,806 square pixels; p=0.013) and bone content (mean bone volume at 20 days postfracture, 11.68±3.18 mm3 versus 22.34±10.59 mm3; p<0.001) compared with the young adult mice. However, the amount of cartilage and bone relative to the total callus size was similar between the adult and geriatric mice (mean bone volume fraction at 25 days postfracture, 0.48±0.10 versus 0.50±0.13; p=0.793), and the relative expression of chondrogenic (mean fold change in SOX9 at 10 days postfracture, 135+25 versus 90±52; p=0.221) and osteogenic genes (mean fold change in osterix at 20 days postfracture, 22.2±5.3 versus 18.7±5.2; p=0.324) was similar. Analysis of mesenchymal cell proliferation in the geriatric mice relative to adult mice showed a decrease in proliferation (mean percent of undifferentiated mesenchymal cells staining proliferating cell nuclear antigen [PCNA] positive at 10 days postfracture, 25%±6.8% versus 42%±14.5%; p=0.047). CONCLUSIONS Our findings suggest that the molecular program of fracture healing is intact in geriatric mice, as it is in geriatric humans, but callus expansion is reduced in magnitude. CLINICAL RELEVANCE Our study showed altered healing capacity in a relevant animal model of geriatric fracture healing. The understanding that callus expansion and bone volume are decreased with aging can help guide the development of targeted therapeutics for these difficult to heal fractures.
Collapse
Affiliation(s)
- Luke A. Lopas
- />Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 2 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104 USA
| | - Nicole S. Belkin
- />Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 2 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104 USA
| | - Patricia L. Mutyaba
- />Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 2 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104 USA
- />Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, Kennett Square, PA USA
| | - Chancellor F. Gray
- />Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 2 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104 USA
| | - Kurt D. Hankenson
- />Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 2 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104 USA
- />Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, Kennett Square, PA USA
| | - Jaimo Ahn
- />Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 2 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104 USA
| |
Collapse
|
29
|
Ode A, Duda GN, Geissler S, Pauly S, Ode JE, Perka C, Strube P. Interaction of age and mechanical stability on bone defect healing: an early transcriptional analysis of fracture hematoma in rat. PLoS One 2014; 9:e106462. [PMID: 25187955 PMCID: PMC4154721 DOI: 10.1371/journal.pone.0106462] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 08/07/2014] [Indexed: 12/03/2022] Open
Abstract
Among other stressors, age and mechanical constraints significantly influence regeneration cascades in bone healing. Here, our aim was to identify genes and, through their functional annotation, related biological processes that are influenced by an interaction between the effects of mechanical fixation stability and age. Therefore, at day three post-osteotomy, chip-based whole-genome gene expression analyses of fracture hematoma tissue were performed for four groups of Sprague-Dawley rats with a 1.5-mm osteotomy gap in the femora with varying age (12 vs. 52 weeks - biologically challenging) and external fixator stiffness (mechanically challenging). From 31099 analysed genes, 1103 genes were differentially expressed between the six possible combinations of the four groups and from those 144 genes were identified as statistically significantly influenced by the interaction between age and fixation stability. Functional annotation of these differentially expressed genes revealed an association with extracellular space, cell migration or vasculature development. The chip-based whole-genome gene expression data was validated by q-RT-PCR at days three and seven post-osteotomy for MMP-9 and MMP-13, members of the mechanosensitive matrix metalloproteinase family and key players in cell migration and angiogenesis. Furthermore, we observed an interaction of age and mechanical stimuli in vitro on cell migration of mesenchymal stromal cells. These cells are a subpopulation of the fracture hematoma and are known to be key players in bone regeneration. In summary, these data correspond to and might explain our previously described biomechanical healing outcome after six weeks in response to fixation stiffness variation. In conclusion, our data highlight the importance of analysing the influence of risk factors of fracture healing (e.g. advanced age, suboptimal fixator stability) in combination rather than alone.
Collapse
Affiliation(s)
- Andrea Ode
- Julius Wolff Institute, Charité - Universitätsmedizin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute, Charité - Universitätsmedizin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
- Klinik für Orthopädie, Centrum für Muskuloskeletale Chirurgie, Charité - Universitätsmedizin, Berlin, Germany
- * E-mail:
| | - Sven Geissler
- Julius Wolff Institute, Charité - Universitätsmedizin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Stephan Pauly
- Julius Wolff Institute, Charité - Universitätsmedizin, Berlin, Germany
- Klinik für Orthopädie, Centrum für Muskuloskeletale Chirurgie, Charité - Universitätsmedizin, Berlin, Germany
| | - Jan-Erik Ode
- Julius Wolff Institute, Charité - Universitätsmedizin, Berlin, Germany
| | - Carsten Perka
- Julius Wolff Institute, Charité - Universitätsmedizin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
- Klinik für Orthopädie, Centrum für Muskuloskeletale Chirurgie, Charité - Universitätsmedizin, Berlin, Germany
| | - Patrick Strube
- Julius Wolff Institute, Charité - Universitätsmedizin, Berlin, Germany
- Klinik für Orthopädie, Centrum für Muskuloskeletale Chirurgie, Charité - Universitätsmedizin, Berlin, Germany
| |
Collapse
|
30
|
Yukata K, Xie C, Li TF, Takahata M, Hoak D, Kondabolu S, Zhang X, Awad HA, Schwarz EM, Beck CA, Jonason JH, O'Keefe RJ. Aging periosteal progenitor cells have reduced regenerative responsiveness to bone injury and to the anabolic actions of PTH 1-34 treatment. Bone 2014; 62:79-89. [PMID: 24530870 PMCID: PMC4085793 DOI: 10.1016/j.bone.2014.02.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/17/2014] [Accepted: 02/04/2014] [Indexed: 12/20/2022]
Abstract
A stabilized tibia fracture model was used in young (8-week old) and aged (1-year old) mice to define the relative bone regenerative potential and the relative responsiveness of the periosteal progenitor population with aging and PTH 1-34 (PTH) systemic therapy. Bone regeneration was assessed through gene expressions, radiographic imaging, histology/histomorphometry, and biomechanical testing. Radiographs and microCT showed increased calcified callus tissue and enhanced bone healing in young compared to aged mice. A key mechanism involved reduced proliferation, expansion, and differentiation of periosteal progenitor cell populations in aged mice. The experiments showed that PTH increased calcified callus tissue and torsional strength with a greater response in young mice. Histology and quantitative histomorphometry confirmed that PTH increased callus tissue area due primarily to an increase in bone formation, since minimal changes in cartilage and mesenchyme tissue area occurred. Periosteum examined at 3, 5, and 7 days showed that PTH increased cyclin D1 expression, the total number of cells in the periosteum, and width of the periosteal regenerative tissue. Gene expression showed that aging delayed differentiation of both bone and cartilage tissues during fracture healing. PTH resulted in sustained Col10a1 expression consistent with delayed chondrocyte maturation, but otherwise minimally altered cartilage gene expression. In contrast, PTH 1-34 stimulated expression of Runx2 and Osterix, but resulted in reduced Osteocalcin. β-Catenin staining was present in mesenchymal chondroprogenitors and chondrocytes in early fracture healing, but was most intense in osteoblastic cells at later times. PTH increased active β-catenin staining in the osteoblast populations of both young and aged mice, but had a lesser effect in cartilage. Altogether the findings show that reduced fracture healing in aging involves decreased proliferation and differentiation of stem cells lining the bone surface. While PTH 1-34 enhances the proliferation and expansion of the periosteal stem cell population and accelerates bone formation and fracture healing, the effects are proportionately reduced in aged mice compared to young mice. β-Catenin is induced by PTH in early and late fracture healing and is a potential target of PTH 1-34 effects.
Collapse
Affiliation(s)
- Kiminori Yukata
- Department of Orthopedics, Tokushima University Hospital, Kuramoto, Tokushima, Japan.
| | - Chao Xie
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Tian-Fang Li
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Masahiko Takahata
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Donna Hoak
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Sirish Kondabolu
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Xinping Zhang
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Hani A Awad
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Edward M Schwarz
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Christopher A Beck
- Department of Biostatistics and Computational Biology, University of Rochester, USA.
| | - Jennifer H Jonason
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| | - Regis J O'Keefe
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
31
|
Wang W, Shen H, Xie J, Zhou Q, Chen Y, Lu H. Bioinformatics analysis of time-series genes profiling to explore key genes affected by age in fracture healing. Mol Biol Rep 2014; 41:3881-9. [PMID: 24627361 DOI: 10.1007/s11033-014-3255-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/08/2014] [Indexed: 01/23/2023]
Abstract
The present study was aimed to explore possible key genes and bioprocess affected by age during fracture healing. GSE589, GSE592 and GSE1371 were downloaded from gene expression omnibus database. The time-series genes of three age levels rats were firstly identified with hclust function in R. Then functional and pathway enrichment analysis for selected time-series genes were performed. Finally, the VennDiagram package of R language was used to screen overlapping n time-series genes. The expression changes of time-series genes in the rats of three age levels were classified into two types: one was higher expressed at 0 day, decreased at 3 day to 2 week, and increased from 4 to 6 week; the other was the opposite. Functional and pathways enrichment analysis showed that 12 time-series genes of adult and old rats were significantly involved in ECM-receptor interaction pathway. The expression changes of 11 genes were consistent with time axis, 10 genes were up-regulated at 3 days after fracture, and increased slowly in 6 week, while Itga2b was down-regulated. The functions of 106 overlapping genes were all associated with growth and development of bone after fracture. The key genes in ECM-receptor interaction pathway including Spp1, Ibsp, Tnn and Col3a1 have been reported to be related to fracture in literatures. The difference during fracture healing in three age levels rats is mainly related to age. The Spp1, Ibsp, Tnn and Col3a1 are possible potential age-related genes and ECM-receptor interaction pathway is the potential age-related process during fracture healing.
Collapse
Affiliation(s)
- Wei Wang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
The most common procedure that has been developed for use in rats and mice to model fracture healing is described. The nature of the regenerative processes that may be assessed and the types of research questions that may be addressed with this model are briefly outlined. The detailed surgical protocol to generate closed simple transverse fractures is presented, and general considerations when setting up an experiment using this model are described.
Collapse
|
33
|
Komrakova M, Sehmisch S, Tezval M, Ammon J, Lieberwirth P, Sauerhoff C, Trautmann L, Wicke M, Dullin C, Stuermer KM, Stuermer EK. Identification of a vibration regime favorable for bone healing and muscle in estrogen-deficient rats. Calcif Tissue Int 2013; 92:509-20. [PMID: 23416966 PMCID: PMC3654182 DOI: 10.1007/s00223-013-9706-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 01/21/2013] [Indexed: 11/30/2022]
Abstract
Numerous whole-body vibration (WBV) devices of various forces are available on the market, although their influence on the musculoskeletal system is not yet understood. The effect of different WBVs on bone healing and muscle function was evaluated in rats ovariectomized at 3 months of age. 2 months after ovariectomy, bilateral metaphyseal tibia osteotomy and T-plate osteosynthesis were performed. Rats were divided into groups: intact, OVX, and OVX exposed to vertical WBVs of 35, 50, 70, or 90 Hz (experiment 1) or horizontal WBVs of 30, 50, 70, or 90 Hz (experiment 2) 5 days after osteotomy (0.5 mm, 15 min/day for 30 days). The tibia and gastrocnemius and soleus muscles were collected. Vertical vibrations (>35 Hz) improved cortical and callus densities, enlarged callus area and width, suppressed the tartrate-resistant acid phosphatase gene, enhanced citrate synthase activity, accelerated osteotomy bridging (35 and 50 Hz), upregulated the osteocalcin (Oc) gene (70 Hz), and increased relative muscle weight (50 Hz). Horizontal vibrations reduced cortical width (<90 Hz) and callus density (30 Hz), enhanced alkaline phosphatase (Alp) gene expression (50 Hz), decreased the size of oxidative fibers (35 and 70 Hz), and increased capillary density (70, 90 Hz). Biomechanical data; serum Oc, Alp, and creatine kinase activities; body weight; and food intake did not change after WBVs. Vertical WBVs of 35 and 50 Hz produced more favorable results than the higher frequencies. Horizontal WBV showed no positive or negative effects. Further studies are needed to elucidate the effects of WBV on different physiological systems, and precautions must be taken when implementing WBV in the treatment of patients.
Collapse
Affiliation(s)
- Marina Komrakova
- Department of Trauma Surgery and Reconstructive Surgery, University Medical Center Goettingen, Robert-Koch 40, 37075, Goettingen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gene expression signature of human HepG2 cell line. Gene 2013; 518:335-45. [DOI: 10.1016/j.gene.2012.12.106] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 12/21/2012] [Accepted: 12/24/2012] [Indexed: 01/12/2023]
|
35
|
Lau KHW, Kothari V, Das A, Zhang XB, Baylink DJ. Cellular and molecular mechanisms of accelerated fracture healing by COX2 gene therapy: studies in a mouse model of multiple fractures. Bone 2013; 53:369-81. [PMID: 23314071 DOI: 10.1016/j.bone.2013.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/31/2012] [Accepted: 01/02/2013] [Indexed: 01/14/2023]
Abstract
This study sought to determine the cellular and molecular mechanisms of cyclooxygenase-2 (COX2) gene therapy to accelerate fracture repair in a mouse multiple tibial fractures model. The lenti-COX2 (or lenti-gfp control vector) was injected into fractures on day 1 post-fracture. At days 3-7, the COX2 treatment increased Sdf1-, Cxcr4-, Nes-, and Podxl-expressing mesenchymal stem cells (MSCs) within fracture calluses, suggesting an enhanced MSC recruitment or expansion. The COX2-treated mice formed smaller cartilaginous calluses that had less cartilage tissues than control mice. The expression of Sox9 mRNA was 7-fold less in COX2-treated than in control calluses at day 14, implying that COX2 reduces chondrocytic differentiation of MSCs. The therapy also enhanced angiogenesis as reflected by increased immunostaining of CD31, vWF, and α-SMA over controls in the cartilaginous callus at day 14-21. At which time, the COX2 gene therapy promoted bony remodeling of the cartilaginous callus to bridge the fracture gap that was accompanied by 2-fold increase in osteoclasts along the surface of the woven bone and an onset of osteogenesis. Blocking angiogenesis with daily injection of endostatin from day 4 to day 10 into fracture sites blocked the COX2-mediated reduction of callus size that was associated with an increase in hypertrophic chondrocytes and concomitant reduction in osteoclasts. In conclusion, COX2 accelerates fracture healing in part through three biological actions: 1) increased recruitment/expansion of MSCs; 2) decreased cartilaginous callus formation; and 3) increased angiogenesis-dependent cartilage remodeling. These effects were associated with an earlier onset of bony bridging of the fracture gap.
Collapse
Affiliation(s)
- K-H William Lau
- Division of Regenerative Medicine, Loma Linda University School of Medicine, Department of Medicine, Loma Linda, CA92350, USA.
| | | | | | | | | |
Collapse
|
36
|
SURGICAL AND MEDICAL MANAGEMENT FOR FRACTURES OF THE SECOND THROUGH FIFTH METACARPALS IN A RED RUFFED LEMUR (VARECIA RUBRA). J Zoo Wildl Med 2013; 44:215-9. [DOI: 10.1638/1042-7260-44.1.215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
37
|
Abstract
Bone healing after fracture occurs in a well-organized manner and involves a multitude of cell types, inflammatory cytokines, growth factors, prostaglandins, and certain vitamins. Some of the means by which alterations in these essential components affect bone repair are understood, whereas others still need to be delineated. Based on clinical experience and basic science research, certain clinical conditions have become associated with delays in bone repair after fracture. These conditions include chronic inflammation, diabetes, hypovitaminosis, aging, and polytrauma. This brief report reviews some of the ways by which these conditions have been shown to negatively influence bone repair.
Collapse
|
38
|
Thevendran G, Younger A, Pinney S. Current concepts review: risk factors for nonunions in foot and ankle arthrodeses. Foot Ankle Int 2012; 33:1031-40. [PMID: 23131455 DOI: 10.3113/fai.2012.1031] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
39
|
Nakanishi T, Kokubun K, Oda H, Aoki M, Soma A, Taniguchi M, Kazuki Y, Oshimura M, Sato K. Bioluminescence imaging of bone formation using hairless osteocalcin-luciferase transgenic mice. Bone 2012; 51:369-75. [PMID: 22732553 DOI: 10.1016/j.bone.2012.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 05/20/2012] [Accepted: 06/15/2012] [Indexed: 11/19/2022]
Abstract
Osteocalcin is a major noncollagenous protein component of bone extracellular matrix, synthesized and secreted exclusively by osteoblastic cells during the late stage of maturation. We introduced a 10kb human osteocalcin enhancer/promoter (OC)-luciferase (Luc) construct into a hairless mouse line. Examination of tissue RNAs from these transgenic mice showed a predominant restriction of Luc mRNA expression to bone-associated tissues. Immunohistochemical staining of calvaria tissue sections revealed the localization of Luc protein to osteoblasts. Utilizing in vivo bioluminescence imaging, supplementation of 1α,25-dihydroxyvitamin D(3) increased Luc activity throughout the skeleton, consistent with in vitro transient transfection studies in osteoblast-like cells. Moreover, we observed an abrupt decrease in bioluminescence activity as the mice reached puberty, and a further decrease gradually thereafter. Using a radius skeletal repair model, we observed enhanced bioluminescence at the fracture site in both young (14-22 weeks old) and aged (50-66 weeks old) mice. However, peak bioluminescence was delayed in aged mice compared with young mice, suggesting retarded osteocalcin expression with aging. Our in vivo imaging system may contribute to the therapy and prevention of various bone metabolic disorders through its effective monitoring of the bone formation process.
Collapse
Affiliation(s)
- Tomoko Nakanishi
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
This review is aimed at clinicians appraising preclinical trauma studies and researchers investigating compromised bone healing or novel treatments for fractures. It categorises the clinical scenarios of poor healing of fractures and attempts to match them with the appropriate animal models in the literature. We performed an extensive literature search of animal models of long bone fracture repair/nonunion and grouped the resulting studies according to the clinical scenario they were attempting to reflect; we then scrutinised them for their reliability and accuracy in reproducing that clinical scenario. Models for normal fracture repair (primary and secondary), delayed union, nonunion (atrophic and hypertrophic), segmental defects and fractures at risk of impaired healing were identified. Their accuracy in reflecting the clinical scenario ranged greatly and the reliability of reproducing the scenario ranged from 100% to 40%. It is vital to know the limitations and success of each model when considering its application.
Collapse
Affiliation(s)
- L. A. Mills
- Royal National Orthopaedic Hospital, Stanmore, Brockley
Hill, Middlesex HA7 4LP, UK
| | - A. H. R. W. Simpson
- Edinburgh University, Department
of Orthopaedics and Trauma, Chancellors Building, Little
France, Edinburgh EH16 4SB, UK
| |
Collapse
|
41
|
Is the expression of Transforming Growth Factor-Beta1 after fracture of long bones solely influenced by the healing process? INTERNATIONAL ORTHOPAEDICS 2012; 36:2173-9. [PMID: 22623064 DOI: 10.1007/s00264-012-1575-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 05/03/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE Circulating TGF-β1 levels were found to be a predictor of delayed bone healing and non-union. We therefore aimed to investigate some factors that can influence the expression of TGF-β1. The correlation between the expression of TGF-β1 and the different socio-demographic parameters was analysed. METHODS Fifty-one patients with long bone fractures were included in the study and divided into different groups according to their age, gender, cigarette smoking status, diabetes mellitus and regular alcohol intake. TGF-β1 levels were analysed in patient's serum and different groups were retrospectively compared. RESULTS Significantly lower TFG-β1 serum concentrations were observed in non-smokers compared to smokers at week 8 after surgery. Significantly higher concentrations were found in male patients compared to females at week 24. Younger patients had significantly higher concentrations at week 24 after surgery compared to older patients. Concentrations were significantly higher in patients without diabetes compared to those with diabetes at six weeks after surgery. Patients with chronic alcohol abuse had significantly higher concentrations compared to those patients without chronic alcohol abuse. CONCLUSION TGF-β1 serum concentrations vary depending upon smoking status, age, gender, diabetes mellitus and chronic alcohol abuse at different times and therefore do not seem to be a reliable predictive marker as a single-point-in-time measurement for fracture healing.
Collapse
|
42
|
D'Amelio P, Roato I, D'Amico L, Veneziano L, Suman E, Sassi F, Bisignano G, Ferracini R, Gargiulo G, Castoldi F, Pescarmona GP, Isaia GC. Bone and bone marrow pro-osteoclastogenic cytokines are up-regulated in osteoporosis fragility fractures. Osteoporos Int 2011; 22:2869-77. [PMID: 21116815 DOI: 10.1007/s00198-010-1496-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 10/29/2010] [Indexed: 12/21/2022]
Abstract
UNLABELLED This study evaluates cytokines production in bone and bone marrow of patients with an osteoporotic fracture or with osteoarthritis by real time PCR, Western blot and immunohistochemistry. We demonstrate that the cytokine pattern is shifted towards osteoclast activation and osteoblast inhibition in patients with osteoporotic fractures. INTRODUCTION Fragility fractures are the resultant of low bone mass and poor bone architecture typical of osteoporosis. Cytokines involved in the control of bone cell maturation and function are produced by both bone itself and bone marrow cells, but the roles of these two sources in its control and the amounts they produce are not clear. This study compares their production in patients with an osteoporotic fracture and those with osteoarthritis. METHODS We evaluated 52 femoral heads from women subjected to hip-joint replacement surgery for femoral neck fractures due to low-energy trauma (37), or for osteoarthritis (15). Total RNA was extracted from both bone and bone marrow, and quantitative PCR was used to identify the receptor activator of nuclear factor kB Ligand (RANKL), osteoprotegerin (OPG), macrophage colony stimulating factor (M-CSF), transforming growth factor β (TGFβ), Dickoppf-1 (DKK-1) and sclerostin (SOST) expression. Immunohistochemistry and Western blot were performed in order to quantify and localize in bone and bone marrow the cytokines. RESULTS We found an increase of RANKL/OPG ratio, M-CSF, SOST and DKK-1 in fractured patients, whereas TGFβ was increased in osteoarthritic bone. Bone marrow produced greater amounts of RANKL, M-CSF and TGFβ compared to bone, whereas the production of DKK-1 and SOST was higher in bone. CONCLUSIONS We show that bone marrow cells produced the greater amount of pro-osteoclastogenic cytokines, whereas bone cells produced higher amount of osteoblast inhibitors in patients with fragility fracture, thus the cytokine pattern is shifted towards osteoclast activation and osteoblast inhibition in these patients.
Collapse
Affiliation(s)
- P D'Amelio
- Gerontology Section, Department of Surgical and Medical Disciplines, University of Torino, Corso Bramante 88/90, Torino, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Park KH, Kang JW, Lee EM, Kim JS, Rhee YH, Kim M, Jeong SJ, Park YG, Kim SH. Melatonin promotes osteoblastic differentiation through the BMP/ERK/Wnt signaling pathways. J Pineal Res 2011; 51:187-94. [PMID: 21470302 DOI: 10.1111/j.1600-079x.2011.00875.x] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although melatonin has a variety of biological actions such as antitumor, antiangiogenic, and antioxidant activities, the osteogenic mechanism of melatonin still remains unclear. Thus, in the present study, the molecular mechanism of melatonin was elucidated in the differentiation of mouse osteoblastic MC3T3-E1 cells. Melatonin enhanced osteoblastic differentiation and mineralization compared to untreated controls in preosteoblastic MC3T3-E1 cells. Also, melatonin increased wound healing and dose-dependently activated osteogenesis markers such as runt-related transcription factor 2 (Runx2), osteocalcin (OCN), bone morphogenic protein (BMP)-2 and -4 in MC3T3-E1 cells. Of note, melatonin activated Wnt 5 α/β, β-catenin and the phosphorylation of c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) in a time-dependent manner while it attenuated phosphorylation of glycogen synthase kinase 3 beta (GSK-3β) in MC3T3-E1 cells. Consistently, confocal microscope observation revealed that BMP inhibitor Noggin blocked melatonin-induced nuclear localization of β-catenin. Furthermore, Western blotting showed that Noggin reversed activation of β-catenin and Wnt5 α/β and suppression of GSK-3β induced by melatonin in MC3T3-E1 cells, which was similarly induced by ERK inhibitor PD98059. Overall, these findings demonstrate that melatonin promotes osteoblastic differentiation and mineralization in MC3T3-E1 cells via the BMP/ERK/Wnt pathways.
Collapse
Affiliation(s)
- Ki-Ho Park
- Department of Orthodondritics, Kyung-Hee University College of Dental Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jäger M, Ott CE, Grünhagen J, Hecht J, Schell H, Mundlos S, Duda GN, Robinson PN, Lienau J. Composite transcriptome assembly of RNA-seq data in a sheep model for delayed bone healing. BMC Genomics 2011; 12:158. [PMID: 21435219 PMCID: PMC3074554 DOI: 10.1186/1471-2164-12-158] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 03/24/2011] [Indexed: 11/28/2022] Open
Abstract
Background The sheep is an important model organism for many types of medically relevant research, but molecular genetic experiments in the sheep have been limited by the lack of knowledge about ovine gene sequences. Results Prior to our study, mRNA sequences for only 1,556 partial or complete ovine genes were publicly available. Therefore, we developed a composite de novo transcriptome assembly method for next-generation sequence data to combine known ovine mRNA and EST sequences, mRNA sequences from mouse and cow, and sequences assembled de novo from short read RNA-Seq data into a composite reference transcriptome, and identified transcripts from over 12 thousand previously undescribed ovine genes. Gene expression analysis based on these data revealed substantially different expression profiles in standard versus delayed bone healing in an ovine tibial osteotomy model. Hundreds of transcripts were differentially expressed between standard and delayed healing and between the time points of the standard and delayed healing groups. We used the sheep sequences to design quantitative RT-PCR assays with which we validated the differential expression of 26 genes that had been identified by RNA-seq analysis. A number of clusters of characteristic expression profiles could be identified, some of which showed striking differences between the standard and delayed healing groups. Gene Ontology (GO) analysis showed that the differentially expressed genes were enriched in terms including extracellular matrix, cartilage development, contractile fiber, and chemokine activity. Conclusions Our results provide a first atlas of gene expression profiles and differentially expressed genes in standard and delayed bone healing in a large-animal model and provide a number of clues as to the shifts in gene expression that underlie delayed bone healing. In the course of our study, we identified transcripts of 13,987 ovine genes, including 12,431 genes for which no sequence information was previously available. This information will provide a basis for future molecular research involving the sheep as a model organism.
Collapse
Affiliation(s)
- Marten Jäger
- Institute for Medical Genetics, Charité-Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
KATANO M, NARUSE K, UCHIDA K, MIKUNI-TAKAGAKI Y, TAKASO M, ITOMAN M, URABE K. Low Intensity Pulsed Ultrasound Accelerates Delayed Healing Process by Reducing the Time Required for the Completion of Endochondral Ossification in the Aged Mouse Femur Fracture Model. Exp Anim 2011; 60:385-95. [DOI: 10.1538/expanim.60.385] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Motoaki KATANO
- Departments of Orthopedic Surgery, Kitasato University School of Medicine
| | - Kouji NARUSE
- Departments of Orthopedic Surgery, Kitasato University School of Medicine
| | - Kentaroo UCHIDA
- Departments of Orthopedic Surgery, Kitasato University School of Medicine
| | - Yuko MIKUNI-TAKAGAKI
- Department of Maxillofacial Diagnostic Science and Functional Biology, Kanagawa Dental College
| | - Masashi TAKASO
- Departments of Orthopedic Surgery, Kitasato University School of Medicine
| | | | - Ken URABE
- Departments of Orthopedic Surgery, Kitasato University School of Medicine
| |
Collapse
|
46
|
Kopf S, Schenkengel JP, Wieners G, Stärke C, Becker R. No bone tunnel enlargement in patients with open growth plates after transphyseal ACL reconstruction. Knee Surg Sports Traumatol Arthrosc 2010; 18:1445-51. [PMID: 20127314 DOI: 10.1007/s00167-009-1041-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 12/26/2009] [Indexed: 01/11/2023]
Abstract
Bone tunnel enlargement after ACL reconstruction has been described extensively in adults. However, little is known about this phenomenon in patients with open growth plates. Thus, the goals of the current study were to evaluate changes in bone tunnel size in patients with open growth plates after transphyseal ACL reconstruction with suspensory fixation and to correlate tunnel size with clinical outcome after medium-term follow-up. Fourteen patients with open growth plates were included that underwent primary transphyseal ACL reconstruction using hamstrings autografts and suspensory fixation. Mean follow-up time was 7 years. At the time of follow-up, MRIs of the operated knee were performed, and outcome was assessed using KOS-ADLS, Lysholm score, IKDC Subjective Knee Form score, Knee Examination Form score, and KT-1000 measurements. On MRI, the cross-sectional area of the bone tunnels was assessed using special axial cuts perpendicular to the axes of the tunnels. Two orthopaedic surgeons and two radiologists analysed the MRIs. Change in bone tunnel size from surgery to follow-up was calculated. No significant changes in bone tunnel size from surgery to follow-up were found. Regarding outcome measures, KOS-ADLS averaged 95%, Lysholm Score averaged 96 points, IKDC Subjective Knee Form averaged 95%, IKDC Knee Examination Form scores were 8A, 5B, 1C, and KT-1000 measurements averaged 1.8 ± 1.4 mm. No significant correlations were found between tunnel size at follow-up and outcome measures. Based on our study, bone tunnel enlargement does not occur in patients who have open growth plates and undergo ACL reconstruction using suspensory fixation.
Collapse
Affiliation(s)
- S Kopf
- Department of Orthopaedic Surgery, Otto-von-Guericke University of Magdeburg, Magdeburg, Germany.
| | | | | | | | | |
Collapse
|
47
|
Wang Q, Huang C, Zeng F, Xue M, Zhang X. Activation of the Hh pathway in periosteum-derived mesenchymal stem cells induces bone formation in vivo: implication for postnatal bone repair. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:3100-11. [PMID: 20971735 DOI: 10.2353/ajpath.2010.100060] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
While the essential role of periosteum in cortical bone repair and regeneration is well established, the molecular pathways that control the early osteogenic and chondrogenic differentiation of periosteal stem/progenitor cells during repair processes are unclear. Using a murine segmental bone graft transplantation model, we isolated a population of early periosteum-callus-derived mesenchymal stem cells (PCDSCs) from the healing autograft periosteum. These cells express typical mesenchymal stem cell markers and are capable of differentiating into osteoblasts, adipocytes, and chondrocytes. Characterization of these cells demonstrated that activation of the hedgehog (Hh) pathway effectively promoted osteogenic and chondrogenic differentiation of PCDSCs in vitro and induced bone formation in vivo. To determine the role of the Hh pathway in adult bone repair, we deleted Smoothened (Smo), the receptor that transduces all Hh signals at the onset of bone autograft repair via a tamoxifen-inducible RosaCreER mouse model. We found that deletion of Smo markedly reduced osteogenic differentiation of isolated PCDSCs and further resulted in a near 50% reduction in periosteal bone callus formation at the cortical bone junction as determined by MicroCT and histomorphometric analyses. These data strongly suggest that the Hh pathway plays an important role in adult bone repair via enhancing differentiation of periosteal progenitors and that activation of the Hh pathway at the onset of healing could be beneficial for repair and regeneration.
Collapse
Affiliation(s)
- Qun Wang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | | | |
Collapse
|
48
|
Komrakova M, Stuermer EK, Werner C, Wicke M, Kolios L, Sehmisch S, Tezval M, Daub F, Martens T, Witzenhausen P, Dullin C, Stuermer KM. Effect of human parathyroid hormone hPTH (1-34) applied at different regimes on fracture healing and muscle in ovariectomized and healthy rats. Bone 2010; 47:480-92. [PMID: 20580683 DOI: 10.1016/j.bone.2010.05.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Revised: 05/03/2010] [Accepted: 05/10/2010] [Indexed: 10/19/2022]
Abstract
Three experiments were conducted to investigate the effect of intermittent administration of parathyroid hormone (PTH) (1-34) applied at different regimes on fracture healing and muscle in healthy and ovariectomized (Ovx at 3 months of age) rats. Five-month old rats underwent bilateral transverse metaphyseal osteotomy of tibia and were divided into groups (12 rats each). In Exp 1, Ovx rats were either treated with PTH (7x/w, 1-35d), with oral estradiol-17beta-benzoate (0.4 mg/kg BW, 1-35d) or untreated. In Exp. 2, there were 3 groups: healthy untreated or treated with PTH (5x/w, 1-35d or 7-35d). In Exp. 3, there were 7 groups: healthy, Ovx, "healthy PTH 5x/w 7-35d", "Ovx PTH 5x/w 7-35d, 14-35d or 14-28d", "Ovx PTH every other day 7-35d". Single dosage of PTH was 40 microg/kg BW. After 35 days of healing one tibia was analyzed by computed tomographical, biomechanical, histological analyses. The other tibia was used in analyses of Alp, Oc, Trap 1, Igf-1, Rankl, Opg genes (Exp.2, 3). Serum Oc and Alp were measured. Body, uterus weight was recorded. M. gastrocnemius was analyzed for weight (Exp. 2), fiber size and mitochondrial respiratory activity (MRA) (Exp.3). Estrogen enhanced uterus weight, prevented body increase, however, did not improve bone healing in Ovx rats (Exp. 1). PTH administration from days 1 and 7 improved bone parameters in all rats regardless of the application frequency (7, 5x/w or every other day) (Exp. 1, stiffness Ovx: 118+13 N/mm, Ovx PTH: 250+/-20 N/mm) being more effective in healthy rats (Exp. 3, stiffness improvement Healthy: 59 to 174 N/mm, Ovx: 52 to 98 N/mm). Serum Oc level was elevated in PTH treated rats. Application from day 14 proved to be less effective (Exp. 3). PTH had no effect (P>0.05) on body, uterus and muscle weight, muscle fiber size, MRA and expression of bone markers. PTH promoted bone healing in Ovx and healthy rats, when it is applied during early stage of healing without having any adverse systemic effect. In perspective, PTH may represent a treatment for enhancement of fracture healing. The findings need to be confirmed by follow-up studies on other animals.
Collapse
Affiliation(s)
- Marina Komrakova
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch 40, 37075 Goettingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Xing Z, Lu C, Hu D, Miclau T, Marcucio RS. Rejuvenation of the inflammatory system stimulates fracture repair in aged mice. J Orthop Res 2010; 28:1000-6. [PMID: 20108320 PMCID: PMC2892015 DOI: 10.1002/jor.21087] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 11/16/2009] [Indexed: 02/04/2023]
Abstract
Age significantly reduces the regenerative capacity of the skeleton, but the underlying causes are unknown. Here, we tested whether the functional status of inflammatory cells contributes to delayed healing in aged animals. We created chimeric mice by bone marrow transplantation after lethal irradiation. In this model, chondrocytes and osteoblasts in the regenerate are derived exclusively from host cells while inflammatory cells are derived from the donor. Using this model, the inflammatory system of middle-aged mice (12 month old) was replaced by transplanted bone marrow from juvenile mice (4 weeks old), or age-matched controls. We found that the middle-aged mice receiving juvenile bone marrow had larger calluses and more bone formation during early stages and faster callus remodeling at late stages of fracture healing, indicating that inflammatory cells derived from the juvenile bone marrow accelerated bone repair in the middle-aged animals. In contrast, transplanting bone marrow from middle-aged mice to juvenile mice did not alter the process of fracture healing in juvenile mice. Thus, the roles of inflammatory cells in fracture healing may be age-related, suggesting the possibility of enhancing fracture healing in aged animals by manipulating the inflammatory system.
Collapse
Affiliation(s)
- Zhiqing Xing
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California at San Francisco, 2550 23rd Street, Bldg. 9, Rm. 345, San Francisco, California 94110, USA
| | | | | | | | | |
Collapse
|
50
|
Stuermer EK, Komrakova M, Werner C, Wicke M, Kolios L, Sehmisch S, Tezval M, Utesch C, Mangal O, Zimmer S, Dullin C, Stuermer KM. Musculoskeletal response to whole-body vibration during fracture healing in intact and ovariectomized rats. Calcif Tissue Int 2010; 87:168-80. [PMID: 20532877 PMCID: PMC2903688 DOI: 10.1007/s00223-010-9381-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 05/13/2010] [Indexed: 11/30/2022]
Abstract
This study investigated the effect of vibration on bone healing and muscle in intact and ovariectomized rats. Thirty ovariectomized (at 3 months of age) and 30 intact 5-month old female Sprague-Dawley rats underwent bilateral metaphyseal osteotomy of tibia. Five days later, half of the ovariectomized and of the intact rats were exposed to whole-body vertical vibration (90 Hz, 0.5 mm, 4 x g acceleration) for 15 min twice a day during 30 days. The other animals did not undergo vibration. After decapitation of rats, one tibia was used for computed tomographic, biomechanical, and histological analyses; the other was used for gene expression analyses of alkaline phosphatase (Alp), osteocalcin (Oc), tartrate-resistant acid phosphatase 1, and insulinlike growth factor 1. Serum Alp and Oc were measured. Mitochondrial activity, fiber area and distribution, and capillary densities were analyzed in M. gastrocnemius and M. longissimus. We found that vibration had no effect on body weight and food intake, but it improved cortical and callus densities (97 vs. 99%, 72 vs. 81%), trabecular structure (9 vs. 14 trabecular nodes), blood supply (1.7 vs. 2.1 capillaries/fiber), and oxidative metabolism (17 vs. 23 pmol O(2)/s/mg) in ovariectomized rats. Vibration generally increased muscle fiber size. Tibia biomechanical properties were diminished after vibration. Oc gene expression was higher in vibrated rats. Serum Alp was increased in ovariectomized rats. In ovariectomized rats, vibration resulted in an earlier bridging; in intact rats, callus bridging occurred later after vibration. The chosen vibration regimen (90 Hz, 0.5 mm, 4 x g acceleration, 15 min twice a day) was effective in improving musculoskeletal tissues in ovariectomized rats but was not optimal for fracture healing.
Collapse
Affiliation(s)
- Ewa K. Stuermer
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Marina Komrakova
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Carsten Werner
- Institute of Food Quality and Safety, University of Animal Medicine, Hannover, Germany
| | - Michael Wicke
- Department of Animal Sciences, University of Goettingen, Goettingen, Germany
| | - Leila Kolios
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Stephan Sehmisch
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Mohammad Tezval
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Clara Utesch
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Orzala Mangal
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Sebastian Zimmer
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| | - Christian Dullin
- Department of Radiology, University of Goettingen, Goettingen, Germany
| | - Klaus M. Stuermer
- Department of Trauma Surgery and Reconstructive Surgery, University of Goettingen, Robert-Koch St. 40, 37075 Goettingen, Germany
| |
Collapse
|