1
|
Koh NYY, Miszkiewicz JJ, Fac ML, Wee NKY, Sims NA. Preclinical Rodent Models for Human Bone Disease, Including a Focus on Cortical Bone. Endocr Rev 2024; 45:493-520. [PMID: 38315213 PMCID: PMC11244217 DOI: 10.1210/endrev/bnae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/22/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Preclinical models (typically ovariectomized rats and genetically altered mice) have underpinned much of what we know about skeletal biology. They have been pivotal for developing therapies for osteoporosis and monogenic skeletal conditions, including osteogenesis imperfecta, achondroplasia, hypophosphatasia, and craniodysplasias. Further therapeutic advances, particularly to improve cortical strength, require improved understanding and more rigorous use and reporting. We describe here how trabecular and cortical bone structure develop, are maintained, and degenerate with aging in mice, rats, and humans, and how cortical bone structure is changed in some preclinical models of endocrine conditions (eg, postmenopausal osteoporosis, chronic kidney disease, hyperparathyroidism, diabetes). We provide examples of preclinical models used to identify and test current therapies for osteoporosis, and discuss common concerns raised when comparing rodent preclinical models to the human skeleton. We focus especially on cortical bone, because it differs between small and larger mammals in its organizational structure. We discuss mechanisms common to mouse and human controlling cortical bone strength and structure, including recent examples revealing genetic contributors to cortical porosity and osteocyte network configurations during growth, maturity, and aging. We conclude with guidelines for clear reporting on mouse models with a goal for better consistency in the use and interpretation of these models.
Collapse
Affiliation(s)
- Natalie Y Y Koh
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Justyna J Miszkiewicz
- School of Social Science, The University of Queensland, Brisbane, QLD 4072, Australia
- Vertebrate Evolution Development and Ecology, Naturalis Biodiversity Center, 2333 CR Leiden, The Netherlands
| | - Mary Louise Fac
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie K Y Wee
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie A Sims
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
2
|
Yang G(K, Chen H, Cheng KL, Tang MF, Wang Y, Hung LH(A, Cheng CY(J, Mak KL(K, Lee YW(W. Potential Interaction between WNT16 and Vitamin D on Bone Qualities in Adolescent Idiopathic Scoliosis Patients and Healthy Controls. Biomedicines 2024; 12:250. [PMID: 38275421 PMCID: PMC10813331 DOI: 10.3390/biomedicines12010250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/01/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a three-dimensional spinal deformity that is associated with low bone mineral density (BMD). Vitamin D (Vit-D) supplementation has been suggested to improve BMD in AIS, and its outcomes may be related to genetic factors. The present study aimed to (a) investigate the synergistic effect between a low BMD-related gene (wingless-related integration site 16, WNT16) and two important Vit-D pathway genes (Vit-D receptor, VDR, and Vit-D binding protein, VDBP) on serum Vit-D and bone qualities in Chinese AIS patients and healthy adolescents, and (b) to further investigate the effect of ablating Wnt16 on the cortical bone quality and whether diets with different dosages of Vit-D would further influence bone quality during the rapid growth phase in mice in the absence of Wnt16. A total of 519 girls (318 AIS vs. 201 controls) were recruited, and three selected single-nucleotide polymorphisms (SNPs) (WNT16 rs3801387, VDBP rs2282679, and VDR rs2228570) were genotyped. The serum 25(OH)Vit-D level was significantly associated with VDBP rs2282679 alleles (OR = -4.844; 95% CI, -7.521 to -2.167, p < 0.001). Significant multi-locus models were identified by generalized multifactor dimensionality reduction (GMDR) analyses on the serum 25(OH)Vit-D level (p = 0.006) and trabecular area (p = 0.044). In the gene-edited animal study, Wnt16 global knockout (KO) and wildtype (WT) male mice were provided with different Vit-D diets (control chow (1000 IU/Kg) vs. Vit-D-deficient chow (Nil in Vit-D) vs. high-dose Vit-D chow (20,000 IU/Kg)) from 4 weeks to 10 weeks old. Wnt16 global KO mice had significantly lower serum 25(OH)Vit-D levels and higher liver Vdbp mRNA expression levels than WT mice. In addition, Wnt16 global KO mice showed a decrease in bone density, cortical thickness and cortical area compared with WT mice. Interestingly, high-dose Vit-D chow led to lower bone density, cortical thickness, and cortical area in WT mice, which were less obvious in Wnt16 global KO mice. In conclusion, WNT16 may regulate the serum 25(OH)Vit-D level and bone qualities, which might be associated with VDBP expression. Further investigations with a larger sample size and wider spectrum of scoliosis severity are required to validate our findings regarding the interaction between WNT16 and Vit-D status in patients with AIS.
Collapse
Affiliation(s)
- Guangpu (Kenneth) Yang
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Huanxiong Chen
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Department of Spine Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 571199, China
| | - Ka-Lo Cheng
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Man-Fung Tang
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Yujia Wang
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Lik-Hang (Alec) Hung
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, Hong Kong, China
| | - Chun-Yiu (Jack) Cheng
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Yuk-Wai (Wayne) Lee
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
3
|
Friedman MA, Buettmann EG, Zeineddine Y, Abraham LB, Hoppock GA, Meas SJ, Zhang Y, Farber CR, Donahue HJ. Genetic variation influences the skeletal response to hindlimb unloading in the eight founder strains of the diversity outbred mouse population. J Orthop Res 2024; 42:134-140. [PMID: 37321985 PMCID: PMC10721729 DOI: 10.1002/jor.25646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/11/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
During disuse, mechanical unloading causes extensive bone loss, decreasing bone volume and strength. Variations in bone mass and risk of osteoporosis are influenced by genetics; however, it remains unclear how genetic variation affects the skeletal response to unloading. We previously found that genetic variation affects the musculoskeletal response to 3 weeks of immobilization in the 8 Jackson Laboratory J:DO founder strains: C57Bl/6J, A/J, 129S1/SvImJ, NOD/ShiLtJ, NZO/HlLtJ, CAST/EiJ, PWK/PhJ, and WSB/EiJ. Hindlimb unloading (HLU) is the best model for simulating local and systemic contributors of disuse and therefore may have a greater impact on bones than immobilization. We hypothesized that genetic variation would affect the response to HLU across the eight founder strains. Mice of each founder strain were placed in HLU for 3 weeks, and the femurs and tibias were analyzed. There were significant HLU and mouse strain interactions on body weight, femur trabecular BV/TV, and femur ultimate force. This indicates that unloading only caused significant catabolic effects in some mouse strains. C57BL/6 J mice were most affected by unloading while other strains were more protected. There were significant HLU and mouse strain interactions on gene expression of genes encoding bone metabolism genes in the tibia. This indicates that unloading only caused significant effects on bone metabolism genes in some mouse strains. Different mouse strains respond to HLU differently, and this can be explained by genetic differences. These results suggest the outbred J:DO mice will be a powerful model for examining the effects of genetics on the skeletal response to HLU.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yue Zhang
- Virginia Commonwealth University, Richmond VA
| | | | | |
Collapse
|
4
|
Janssen JN, Kalev-Altman R, Shalit T, Sela-Donenfeld D, Monsonego-Ornan E. Differential gene expression in the calvarial and cortical bone of juvenile female mice. Front Endocrinol (Lausanne) 2023; 14:1127536. [PMID: 37378024 PMCID: PMC10291685 DOI: 10.3389/fendo.2023.1127536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction Both the calvarial and the cortical bones develop through intramembranous ossification, yet they have very different structures and functions. The calvaria enables the rapid while protected growth of the brain, whereas the cortical bone takes part in locomotion. Both types of bones undergo extensive modeling during embryonic and post-natal growth, while bone remodeling is the most dominant process in adults. Their shared formation mechanism and their highly distinct functions raise the fundamental question of how similar or diverse the molecular pathways that act in each bone type are. Methods To answer this question, we aimed to compare the transcriptomes of calvaria and cortices from 21-day old mice by bulk RNA-Seq analysis. Results The results revealed clear differences in expression levels of genes related to bone pathologies, craniosynostosis, mechanical loading and bone-relevant signaling pathways like WNT and IHH, emphasizing the functional differences between these bones. We further discussed the less expected candidate genes and gene sets in the context of bone. Finally, we compared differences between juvenile and mature bone, highlighting commonalities and dissimilarities of gene expression between calvaria and cortices during post-natal bone growth and adult bone remodeling. Discussion Altogether, this study revealed significant differences between the transcriptome of calvaria and cortical bones in juvenile female mice, highlighting the most important pathway mediators for the development and function of two different bone types that originate both through intramembranous ossification.
Collapse
Affiliation(s)
- Jerome Nicolas Janssen
- The Institute of Biochemistry, Food Science and Nutrition, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Rotem Kalev-Altman
- The Institute of Biochemistry, Food Science and Nutrition, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- The Koret School of Veterinary Medicine, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Tali Shalit
- The Ilana and Pascal Mantoux Institute for Bioinformatics, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Dalit Sela-Donenfeld
- The Koret School of Veterinary Medicine, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry, Food Science and Nutrition, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
5
|
Loflin BE, Ahn T, Colglazier KA, Banaszak Holl MM, Ashton-Miller JA, Wojtys EM, Schlecht SH. An Adolescent Murine In Vivo Anterior Cruciate Ligament Overuse Injury Model. Am J Sports Med 2023; 51:1721-1732. [PMID: 37092727 PMCID: PMC10348391 DOI: 10.1177/03635465231165753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
BACKGROUND Overuse ligament and tendon injuries are prevalent among recreational and competitive adolescent athletes. In vitro studies of the ligament and tendon suggest that mechanical overuse musculoskeletal injuries begin with collagen triple-helix unraveling, leading to collagen laxity and matrix damage. However, there are little in vivo data concerning this mechanism or the physiomechanical response to collagen disruption, particularly regarding the anterior cruciate ligament (ACL). PURPOSE To develop and validate a novel in vivo animal model for investigating the physiomechanical response to ACL collagen matrix damage accumulation and propagation in the ACL midsubstance, fibrocartilaginous entheses, and subchondral bone. STUDY DESIGN Controlled laboratory study. METHODS C57BL/6J adolescent inbred mice underwent 3 moderate to strenuous ACL fatigue loading sessions with a 72-hour recovery between sessions. Before each session, randomly selected subsets of mice (n = 12) were euthanized for quantifying collagen matrix damage (percent collagen unraveling) and ACL mechanics (strength and stiffness). This enabled the quasi-longitudinal assessment of collagen matrix damage accrual and whole tissue mechanical property changes across fatigue sessions. Additionally, all cyclic loading data were quantified to evaluate changes in knee mechanics (stiffness and hysteresis) across fatigue sessions. RESULTS Moderate to strenuous fatigue loading across 3 sessions led to a 24% weaker (P = .07) and 35% less stiff (P < .01) ACL compared with nonloaded controls. The unraveled collagen densities within the fatigued ACL and entheseal matrices after the second and third sessions were 38% (P < .01) and 15% (P = .02) higher compared with the nonloaded controls. CONCLUSION This study confirmed the hypothesis that in vivo ACL collagen matrix damage increases with tissue fatigue sessions, adversely impacting ACL mechanical properties. Moreover, the in vivo ACL findings were consistent with in vitro overloading research in humans. CLINICAL RELEVANCE The outcomes from this study support the use of this model for investigating ACL overuse injuries.
Collapse
Affiliation(s)
- Benjamin E. Loflin
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Taeyong Ahn
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kaitlyn A. Colglazier
- Purdue School of Engineering and Technology, Purdue University–Indianapolis, Indianapolis, Indiana, USA
| | - Mark M. Banaszak Holl
- Department of Orthopaedic Surgery, Heersink School of Medicine, University of Alabama–Birmingham, Birmingham, Alabama, USA
| | | | - Edward M. Wojtys
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Stephen H. Schlecht
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Purdue School of Engineering and Technology, Purdue University–Indianapolis, Indianapolis, Indiana, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Genetic variability affects the skeletal response to immobilization in founder strains of the diversity outbred mouse population. Bone Rep 2021; 15:101140. [PMID: 34761080 PMCID: PMC8566767 DOI: 10.1016/j.bonr.2021.101140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Mechanical unloading decreases bone volume and strength. In humans and mice, bone mineral density is highly heritable, and in mice the response to changes in loading varies with genetic background. Thus, genetic variability may affect the response of bone to unloading. As a first step to identify genes involved in bones' response to unloading, we evaluated the effects of unloading in eight inbred mouse strains: C57BL/6J, PWK/PhJ, WSB/EiJ, A/J, 129S1/SvImJ, NOD/ShiLtJ, NZO/HlLtJ, and CAST/EiJ. C57BL/6J and NOD/ShiLtJ mice had the greatest unloading-induced loss of diaphyseal cortical bone volume and strength. NZO/HlLtJ mice had the greatest metaphyseal trabecular bone loss, and C57BL/6J, WSB/EiJ, NOD/ShiLtJ, and CAST/EiJ mice had the greatest epiphyseal trabecular bone loss. Bone loss in the epiphyses displayed the highest heritability. With immobilization, mineral:matrix was reduced, and carbonate:phosphate and crystallinity were increased. A/J mice displayed the greatest unloading-induced loss of mineral:matrix. Changes in gene expression in response to unloading were greatest in NOD/ShiLtJ and CAST/EiJ mice. The most upregulated genes in response to unloading were associated with increased collagen synthesis and extracellular matrix formation. Our results demonstrate a strong differential response to unloading as a function of strain. Diversity outbred (DO) mice are a high-resolution mapping population derived from these eight inbred founder strains. These results suggest DO mice will be highly suited for examining the genetic basis of the skeletal response to unloading. Mouse strain affects bone's response to immobilization. Magnitude of bone loss from immobilization is heritable. Bone transcriptomic response to immobilization is influenced by genetic variation.
Collapse
|
7
|
Li M, Cheng D, Li H, Yao W, Guo D, Wang S, Si J. Tributyltin perturbs femoral cortical architecture and polar moment of inertia in rat. BMC Musculoskelet Disord 2021; 22:427. [PMID: 33962613 PMCID: PMC8106170 DOI: 10.1186/s12891-021-04298-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/20/2021] [Indexed: 01/22/2023] Open
Abstract
Background Tributyltin, a well-known endocrine disruptor, is widely used in agriculture and industry. Previous studies have shown that tributyltin could cause deleterious effects on bone health by impairing the adipo-osteogenic balance in bone marrow. Methods To investigate further the effects of tributyltin on bone, weaned male SD rats were treated with tributyltin (0.5, 5 or 50 μg·kg− 1) or corn oil by gavage once every 3 days for 60 days in this study. Then, we analyzed the effects of tributyltin on geometry, the polar moment of inertia, mineral content, relative abundances of mRNA from representative genes related to adipogenesis and osteogenesis, serum calcium ion and inorganic phosphate levels. Results Micro-computed tomography analysis revealed that treatment with 50 μg·kg− 1 tributyltin caused an obvious decrease in femoral cortical cross sectional area, marrow area, periosteal circumference and derived polar moment of inertia in rats. However, other test results showed that exposure to tributyltin resulted in no significant changes in the expression of genes detected, femoral cancellous architecture, ash content, as well as serum calcium ion and inorganic phosphate levels. Conclusions Exposure to a low dose of tributyltin from the prepubertal to adult stage produced adverse effects on skeletal architecture and strength. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-021-04298-2.
Collapse
Affiliation(s)
- Mingjun Li
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China
| | - Dong Cheng
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Hui Li
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Wenhuan Yao
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Dongmei Guo
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China
| | - Shu'e Wang
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China
| | - Jiliang Si
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China.
| |
Collapse
|
8
|
Jensen VFH, Mølck AM, Dalgaard M, McGuigan FE, Akesson KE. Changes in bone mass associated with obesity and weight loss in humans: Applicability of animal models. Bone 2021; 145:115781. [PMID: 33285255 DOI: 10.1016/j.bone.2020.115781] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/05/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022]
Abstract
The implications of obesity and weight loss for human bone health are not well understood. Although the bone changes associated with weight loss are similar in humans and rodents, that is not the case for obesity. In humans, obesity is generally associated with increased bone mass, an outcome which is exacerbated by advanced age and menopause. In rodents, by contrast, bone mass decreases in proportion to severity and duration of obesity, and is influenced by sex, age and mechanical load. Despite these discrepancies, rodents are frequently used to model the situation in humans. In this review, we summarise the existing knowledge of the effects of obesity and weight loss on bone mass in humans and rodents, focusing on the translatability of findings from animal models. We then describe how animal models should be used to broaden the understanding of the relationship between obesity, weight loss, and skeletal health in humans. Specifically, we highlight the aspects of study design that should be considered to optimise translatability of the rodent models of obesity and weight loss. Notably, the sex, age, and nutritional status of the animals should ideally match those of interest in humans. With these caveats in mind, and depending on the research question asked, our review underscores that animal models can provide valuable information for obesity and weight-management research.
Collapse
Affiliation(s)
- Vivi F H Jensen
- Lund University, Department of Clinical Sciences Malmö and Skåne University Hospital, Department of Orthopedics, Inga Marie Nilssons Gata 22, 205 02 Malmö, Sweden.
| | - Anne-Marie Mølck
- Novo Nordisk A/S, Department of Safety Sciences, Imaging & Data Management, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Majken Dalgaard
- Novo Nordisk A/S, Department of Safety Sciences, Imaging & Data Management, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Fiona E McGuigan
- Lund University, Department of Clinical Sciences Malmö and Skåne University Hospital, Department of Orthopedics, Inga Marie Nilssons Gata 22, 205 02 Malmö, Sweden
| | - Kristina E Akesson
- Lund University, Department of Clinical Sciences Malmö and Skåne University Hospital, Department of Orthopedics, Inga Marie Nilssons Gata 22, 205 02 Malmö, Sweden
| |
Collapse
|
9
|
Schreurs AS, Torres S, Truong T, Moyer EL, Kumar A, Tahimic CGT, Alwood JS, Globus RK. Skeletal tissue regulation by catalase overexpression in mitochondria. Am J Physiol Cell Physiol 2020; 319:C734-C745. [PMID: 32783660 DOI: 10.1152/ajpcell.00068.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Accumulation of oxidative damage from excess reactive oxygen species (ROS) may contribute to skeletal aging and mediate adverse responses to physiological challenges. Wild-type (WT) mice and transgenic mice (male, 16 wk of age) with human catalase targeted to the mitochondria (mCAT) were analyzed for skeletal responses to the remodeling stimuli of combined hind-limb unloading and exposure to ionizing radiation (137Cs, 2 Gy). Treatment for 2 wk caused lipid peroxidation in the bones WT but not mCAT mice, showing that transgene expression mitigated oxidative stress. Ex vivo osteoblast colony growth rate was 95% greater in mCAT than WT mice and correlated with catalase activity levels (P < 0.005, r = 0.67), although terminal osteoblast and osteoclast differentiation were unaffected. mCAT mice had lower cancellous bone volume and cortical size than WT mice. Ambulatory control mCAT animals also displayed reduced cancellous and cortical structural properties compared with control WT mice. In mCAT but not WT mice, treatment caused an unexpectedly rapid radial expansion (+8% cortical area, +22% moment of inertia), reminiscent of compensatory bone growth during advancing age. In contrast, treatment caused similar structural deficits in cancellous tissue of mCAT and WT mice. In sum, mitochondrial ROS signaling via H2O2 was important for the acquisition of adult bone structure and catalase overexpression failed to protect cancellous tissue from treatment. In contrast, catabolic stimuli caused radial expansion in mCAT not WT mice, suggesting that mitochondrial ROS in skeletal cells act to suppress tissue turnover in response to remodeling challenges.
Collapse
Affiliation(s)
- Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Samantha Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Tiffany Truong
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Eric L Moyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Akhhilesh Kumar
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,KBR, Moffett Field, California
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| |
Collapse
|
10
|
King HE, Tommasini SM, Rodriguez-Navarro AB, Mercado BQ, Skinner HCW. Correlative vibrational spectroscopy and 2D X-ray diffraction to probe the mineralization of bone in phosphate-deficient mice. J Appl Crystallogr 2019; 52:960-971. [PMID: 31636517 PMCID: PMC6782074 DOI: 10.1107/s1600576719009361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/29/2019] [Indexed: 11/18/2022] Open
Abstract
Bone crystallite chemistry and structure change during bone maturation. However, these properties of bone can also be affected by limited uptake of the chemical constituents of the mineral by the animal. This makes probing the effect of bone-mineralization-related diseases a complicated task. Here it is shown that the combination of vibrational spectroscopy with two-dimensional X-ray diffraction can provide unparalleled information on the changes in bone chemistry and structure associated with different bone pathologies (phosphate deficiency) and/or health conditions (pregnancy, lactation). Using a synergistic analytical approach, it was possible to trace the effect that changes in the remodelling regime have on the bone mineral chemistry and structure in normal and mineral-deficient (hypophosphatemic) mice. The results indicate that hypophosphatemic mice have increased bone remodelling, increased carbonate content and decreased crystallinity of the bone mineral, as well as increased misalignment of crystallites within the bone tissue. Pregnant and lactating mice that are normal and hypophosphatemic showed changes in the chemistry and misalignment of the apatite crystals that can be related to changes in remodelling rates associated with different calcium demand during pregnancy and lactation.
Collapse
Affiliation(s)
- Helen E King
- Department of Earth Sciences, Utrecht University, Princetonlaan 8a, Utrecht 3584 CB, The Netherlands
- Department of Geology and Geophysics, Yale University, 210 Whitney Avenue, New Haven, Connecticut CT-06511, USA
| | - Steven M Tommasini
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 330 Cedar Street, New Haven, Connecticut CT-06510, USA
| | | | - Brandon Q Mercado
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut CT-06511, USA
| | - H Catherine W Skinner
- Department of Geology and Geophysics, Yale University, 210 Whitney Avenue, New Haven, Connecticut CT-06511, USA
| |
Collapse
|
11
|
Schlecht SH, Martin CT, Ochocki DN, Nolan BT, Wojtys EM, Ashton-Miller JA. Morphology of Mouse Anterior Cruciate Ligament-Complex Changes Following Exercise During Pubertal Growth. J Orthop Res 2019; 37:1910-1919. [PMID: 31042312 PMCID: PMC6700741 DOI: 10.1002/jor.24328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 04/08/2019] [Indexed: 02/04/2023]
Abstract
Postnatal development and the physiological loading response of the anterior cruciate ligament (ACL) complex (ACL proper, entheses, and bony morphology) is not well understood. We tested whether the ACL-complex of two inbred mouse strains that collectively encompass the musculoskeletal variation observed in humans would demonstrate significant morphological differences following voluntary cage-wheel running during puberty compared with normal cage activity controls. Female A/J and C57BL/6J (B6) 6-week-old mice were provided unrestricted access to a standard cage-wheel for 4 weeks. A/J-exercise mice showed a 6.3% narrower ACL (p = 0.64), and a 20.1% more stenotic femoral notch (p < 0.01) while B6-exercise mice showed a 12.3% wider ACL (p = 0.10), compared with their respective controls. Additionally, A/J-exercise mice showed a 5.3% less steep posterior medial tibial slope (p = 0.07) and an 8.8% less steep posterior lateral tibial slope (p = 0.07), while B6-exercise mice showed a 9.8% more steep posterior medial tibial slope (p < 0.01) than their respective controls. A/J-exercise mice also showed more reinforcement of the ACL tibial enthesis with a 20.4% larger area (p < 0.01) of calcified fibrocartilage distributed at a 29.2% greater depth (p = 0.02) within the tibial enthesis, compared with their controls. These outcomes suggest exercise during puberty significantly influences ACL-complex morphology and that inherent morphological differences between these mice, as observed in their less active genetically similar control groups, resulted in a divergent phenotypic outcome between mouse strains. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1910-1919, 2019.
Collapse
Affiliation(s)
- Stephen H. Schlecht
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Colin T. Martin
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan
| | | | - Bonnie T. Nolan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Edward M. Wojtys
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
12
|
Mathis NJ, Adaniya EN, Smith LM, Robling AG, Jepsen KJ, Schlecht SH. Differential changes in bone strength of two inbred mouse strains following administration of a sclerostin-neutralizing antibody during growth. PLoS One 2019; 14:e0214520. [PMID: 30947279 PMCID: PMC6448823 DOI: 10.1371/journal.pone.0214520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/14/2019] [Indexed: 12/02/2022] Open
Abstract
Administration of sclerostin-neutralizing antibody (Scl-Ab) treatment has been shown to elicit an anabolic bone response in growing and adult mice. Prior work characterized the response of individual mouse strains but did not establish whether the impact of Scl-Ab on whole bone strength would vary across different inbred mouse strains. Herein, we tested the hypothesis that two inbred mouse strains (A/J and C57BL/6J (B6)) will show different whole bone strength outcomes following sclerostin-neutralizing antibody (Scl-Ab) treatment during growth (4.5–8.5 weeks of age). Treated B6 femurs showed a significantly greater stiffness (S) (68.8% vs. 46.0%) and maximum load (ML) (84.7% vs. 44.8%) compared to A/J. Although treated A/J and B6 femurs showed greater cortical area (Ct.Ar) similarly relative to their controls (37.7% in A/J and 41.1% in B6), the location of new bone deposition responsible for the greater mass differed between strains and may explain the greater whole bone strength observed in treated B6 mice. A/J femurs showed periosteal expansion and endocortical infilling, while B6 femurs showed periosteal expansion. Post-yield displacement (PYD) was smaller in treated A/J femurs (-61.2%, p < 0.001) resulting in greater brittleness compared to controls; an effect not present in B6 mice. Inter-strain differences in S, ML, and PYD led to divergent changes in work-to-fracture (Work). Work was 27.2% (p = 0.366) lower in treated A/J mice and 66.2% (p < 0.001) greater in treated B6 mice relative to controls. Our data confirmed the anabolic response to Scl-Ab shown by others, and provided evidence suggesting the mechanical benefits of Scl-Ab administration may be modulated by genetic background, with intrinsic growth patterns of these mice guiding the location of new bone deposition. Whether these differential outcomes will persist in adult and elderly mice remains to be determined.
Collapse
Affiliation(s)
- Noah J. Mathis
- School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Emily N. Adaniya
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Lauren M. Smith
- School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Alexander G. Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Karl J. Jepsen
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Stephen H. Schlecht
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
13
|
Schlecht SH, Ramcharan MA, Yang Y, Smith LM, Bigelow EM, Nolan BT, Moss DE, Devlin MJ, Jepsen KJ. Differential Adaptive Response of Growing Bones From Two Female Inbred Mouse Strains to Voluntary Cage-Wheel Running. JBMR Plus 2018; 2:143-153. [PMID: 30283899 PMCID: PMC6124195 DOI: 10.1002/jbm4.10032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/27/2017] [Accepted: 01/04/2018] [Indexed: 11/30/2022] Open
Abstract
The phenotypic response of bones differing in morphological, compositional, and mechanical traits to an increase in loading during growth is not well understood. We tested whether bones of two inbred mouse strains that assemble differing sets of traits to achieve mechanical homeostasis at adulthood would show divergent responses to voluntary cage‐wheel running. Female A/J and C57BL6/J (B6) 4‐week‐old mice were provided unrestricted access to a standard cage‐wheel for 4 weeks. A/J mice have narrow and highly mineralized femora and B6 mice have wide and less mineralized femora. Both strains averaged 2 to 9.5 km of running per day, with the average‐distance run between strains not significantly different (p = 0.133). Exercised A/J femora showed an anabolic response to exercise with the diaphyses showing a 2.8% greater total area (Tt.Ar, p = 0.06) and 4.7% greater cortical area (Ct.Ar, p = 0.012) compared to controls. In contrast, exercised B6 femora showed a 6.2% (p < 0.001) decrease in Tt.Ar (p < 0.001) and a 6.7% decrease in Ct.Ar (p = 0.133) compared to controls, with the femora showing significant marrow infilling (p = 0.002). These divergent morphological responses to exercise, which did not depend on the daily distance run, translated to a 7.9% (p = 0.001) higher maximum load (ML) for exercised A/J femora but no change in ML for exercised B6 femora compared to controls. A consistent response was observed for the humeri but not the vertebral bodies. This differential outcome to exercise has not been previously observed in isolated loading or forced treadmill running regimes. Our findings suggest there are critical factors involved in the metabolic response to exercise during growth that require further consideration to understand how genotype, exercise, bone morphology, and whole‐bone strength interact during growth. © 2018 The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stephen H Schlecht
- Department of Orthopaedic Surgery University of Michigan Ann Arbor MI USA
| | | | | | - Lauren M Smith
- School of Public Health University of Michigan Ann Arbor MI USA
| | - Erin Mr Bigelow
- Department of Orthopaedic Surgery University of Michigan Ann Arbor MI USA
| | - Bonnie T Nolan
- Department of Orthopaedic Surgery University of Michigan Ann Arbor MI USA
| | - Drew E Moss
- Department of Orthopaedic Surgery University of Michigan Ann Arbor MI USA
| | - Maureen J Devlin
- Department of Anthropology University of Michigan Ann Arbor MI USA
| | - Karl J Jepsen
- Department of Orthopaedic Surgery University of Michigan Ann Arbor MI USA
| |
Collapse
|
14
|
Petermann H, Gauthier JA. Fingerprinting snakes: paleontological and paleoecological implications of zygantral growth rings in Serpentes. PeerJ 2018; 6:e4819. [PMID: 29844972 PMCID: PMC5971835 DOI: 10.7717/peerj.4819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 05/01/2018] [Indexed: 11/23/2022] Open
Abstract
We introduce a new non-destructive source of skeletochronological data with applications to species identification, associating disarticulated remains, assessing minimum number of individuals (MNI), and collection management of fossil snakes, but with potential implications for all bony vertebrates, extinct or extant. Study of a diverse sample of Recent henophidian snakes confirms that annual growth cycles (AGCs) visible on the surface of the vertebral zygantrum correspond to lines of arrested growth in osteohistological thin sections and accordingly reflect chronological age. None of the specimens considered here showed signs of remodelling of the zygantrum, suggesting that a complete, unaltered age record is preserved. We tested potential influences on AGCs with a single experimental organism, a male Bogertophis subocularis, that was raised at a controlled temperature and with constant access to mice and water. The conditions in which this individual was maintained, including that it had yet to live through a full reproductive cycle, enabled us to determine that its AGCs reflect only the annual solar cycle, and neither temperature, nor resource availability, nor energy diversion to gametogenesis could explain that it still exhibited lines of arrested growth. Moreover, growth lines in this specimen are deposited toward the end of the growth season in the fall, and not in the winter, during which this individual continued to feed and grow, even though this mid-latitude species would normally be hibernating and not growing. This suggests that growth lines are not caused by hibernation, but reflect the onset of a physiological cycle preparing Bogertophis subocularis for winter rest. That being said, hibernation and reproductive cycle could still influence the amount of time represented by an individual growth line. Growth-line number and AGC spacing-pattern, plus centrum length, are used to estimate MNI of the Early Eocene fossil snake Boavus occidentalis collected from the Willwood Formation over two field seasons during the late 19th century. We identified eight or nine individuals among specimens previously parcelled among two specimen lots collected during those expeditions.
Collapse
Affiliation(s)
- Holger Petermann
- Department of Geology and Geophysics, Yale University, New Haven, CT, United States of America
| | - Jacques A Gauthier
- Department of Geology and Geophysics, Yale University, New Haven, CT, United States of America.,Yale Peabody Museum of Natural History, Yale University, New Haven, CT, United States of America
| |
Collapse
|
15
|
Xin F, Smith LM, Susiarjo M, Bartolomei MS, Jepsen KJ. Endocrine-disrupting chemicals, epigenetics, and skeletal system dysfunction: exploration of links using bisphenol A as a model system. ENVIRONMENTAL EPIGENETICS 2018; 4:dvy002. [PMID: 29732168 PMCID: PMC5920333 DOI: 10.1093/eep/dvy002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 06/08/2023]
Abstract
Early life exposures to endocrine-disrupting chemicals (EDCs) have been associated with physiological changes of endocrine-sensitive tissues throughout postnatal life. Although hormones play a critical role in skeletal growth and maintenance, the effects of prenatal EDC exposure on adult bone health are not well understood. Moreover, studies assessing skeletal changes across multiple generations are limited. In this article, we present previously unpublished data demonstrating dose-, sex-, and generation-specific changes in bone morphology and function in adult mice developmentally exposed to the model estrogenic EDC bisphenol A (BPA) at doses of 10 μg (lower dose) or 10 mg per kg bw/d (upper dose) throughout gestation and lactation. We show that F1 generation adult males, but not females, developmentally exposed to bisphenol A exhibit dose-dependent reductions in outer bone size resulting in compromised bone stiffness and strength. These structural alterations and weaker bone phenotypes in the F1 generation did not persist in the F2 generation. Instead, F2 generation males exhibited greater bone strength. The underlying mechanisms driving the EDC-induced physiological changes remain to be determined. We discuss potential molecular changes that could contribute to the EDC-induced skeletal effects, with an emphasis on epigenetic dysregulation. Furthermore, we assess the necessity of intact sex steroid receptors to mediate these effects. Expanding future assessments of EDC-induced effects to the skeleton may provide much needed insight into one of the many health effects of these chemicals and aid in regulatory decision making regarding exposure of vulnerable populations to these chemicals.
Collapse
Affiliation(s)
- Frances Xin
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren M Smith
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Martha Susiarjo
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY14642, USA
| | - Marisa S Bartolomei
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karl J Jepsen
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Copes LE, Schutz H, Dlugsoz EM, Judex S, Garland T. Locomotor activity, growth hormones, and systemic robusticity: An investigation of cranial vault thickness in mouse lines bred for high endurance running. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 166:442-458. [DOI: 10.1002/ajpa.23446] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 02/02/2018] [Accepted: 02/06/2018] [Indexed: 12/30/2022]
Affiliation(s)
- L. E. Copes
- Department of Medical Sciences, Frank H. Netter MD School of MedicineQuinnipiac UniversityHamden Connecticut06518
| | - H. Schutz
- Department of BiologyPacific Lutheran UniversityTacoma Washington, DC98447
| | - E. M. Dlugsoz
- Department of BiologyUniversity of CaliforniaRiverside, Riverside California92521
| | - S. Judex
- Department of Biomedical EngineeringStony Brook UniversityStony Brook New York11794
| | - T. Garland
- Department of BiologyUniversity of CaliforniaRiverside, Riverside California92521
| |
Collapse
|
17
|
Bailey S, Karsenty G, Gundberg C, Vashishth D. Osteocalcin and osteopontin influence bone morphology and mechanical properties. Ann N Y Acad Sci 2017; 1409:79-84. [PMID: 29044594 PMCID: PMC5730490 DOI: 10.1111/nyas.13470] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/09/2017] [Accepted: 08/18/2017] [Indexed: 01/12/2023]
Abstract
Osteocalcin (OC) and osteopontin (OPN) are major non-collagenous proteins (NCPs) involved in bone matrix organization and deposition. In spite of this, it is currently unknown whether OC and OPN alter bone morphology and consequently affect bone fracture resistance. The goal of this study is to establish the role of OC and OPN in the determination of cortical bone size, shape, and mechanical properties. Our results show that Oc-/- and Opn-/- mice were no different from each other or wild type (WT) with respect to bone morphology (P > 0.1). Bones from mice lacking both NCPs (Oc-/- Opn-/- ) were shorter, with thicker cortices and larger cortical areas, compared with the WT, Oc-/- , and Opn-/- groups (P < 0.05), suggesting a synergistic role for NCPs in the determination of bone morphology. Maximum bending load was significantly different among the groups (P = 0.024), while tissue mineral density and measures of stiffness and strength were not different (P > 0.1). We conclude that the removal of both OC and OPN from bone matrix induces morphological adaptation at the structural level to maintain bone strength.
Collapse
Affiliation(s)
- Stacyann Bailey
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, New York
| | - Caren Gundberg
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut
| | - Deepak Vashishth
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
18
|
Piemontese M, Almeida M, Robling AG, Kim HN, Xiong J, Thostenson JD, Weinstein RS, Manolagas SC, O'Brien CA, Jilka RL. Old age causes de novo intracortical bone remodeling and porosity in mice. JCI Insight 2017; 2:93771. [PMID: 28878136 DOI: 10.1172/jci.insight.93771] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 08/03/2017] [Indexed: 01/08/2023] Open
Abstract
Decreased cortical thickness and increased cortical porosity are the key anatomic changes responsible for osteoporotic fractures in elderly women and men. The cellular basis of these changes is unbalanced endosteal and intracortical osteonal remodeling by the osteoclasts and osteoblasts that comprise the basic multicellular units (BMUs). Like humans, mice lose cortical bone with age, but unlike humans, this loss occurs in the face of sex steroid sufficiency. Mice are therefore an ideal model to dissect age-specific osteoporotic mechanisms. Nevertheless, lack of evidence for endosteal or intracortical remodeling in mice has raised questions about their translational relevance. We show herein that administration of the antiosteoclastogenic cytokine osteoprotegerin to Swiss Webster mice ablated not only osteoclasts, but also endosteal bone formation, demonstrating the occurrence of BMU-based endosteal remodeling. Femoral cortical thickness decreased in aged male and female C57BL/6J mice, as well as F1 hybrids of C57BL/6J and BALB/cBy mice. This decrease was greater in C57BL/6J mice, indicating a genetic influence. Moreover, endosteal remodeling became unbalanced because of increased osteoclast and decreased osteoblast numbers. The porosity of the femoral cortex increased with age but was much higher in females of both strains. Notably, the increased cortical porosity resulted from de novo intracortical remodeling by osteon-like structures. Age-dependent cortical bone loss was associated with increased osteocyte DNA damage, cellular senescence, the senescence-associated secretory phenotype, and increased levels of RANKL. The demonstration of unbalanced endosteal and intracortical remodeling in old mice validates the relevance of this animal model to involutional osteoporosis in humans.
Collapse
Affiliation(s)
- Marilina Piemontese
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Maria Almeida
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ha-Neui Kim
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Jinhu Xiong
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Jeff D Thostenson
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Robert S Weinstein
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Stavros C Manolagas
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Charles A O'Brien
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Robert L Jilka
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
19
|
Meixner CN, Aref MW, Gupta A, McNerny EMB, Brown D, Wallace JM, Allen MR. Raloxifene Improves Bone Mechanical Properties in Mice Previously Treated with Zoledronate. Calcif Tissue Int 2017; 101:75-81. [PMID: 28246928 PMCID: PMC5459622 DOI: 10.1007/s00223-017-0257-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/16/2017] [Indexed: 12/18/2022]
Abstract
Bisphosphonates represent the gold-standard pharmaceutical agent for reducing fracture risk. Long-term treatment with bisphosphonates can result in tissue brittleness which in rare clinical cases manifests as atypical femoral fracture. Although this has led to an increasing call for bisphosphonate cessation, few studies have investigated therapeutic options for follow-up treatment. The goal of this study was to test the hypothesis that treatment with raloxifene, a drug that has cell-independent effects on bone mechanical material properties, could reverse the compromised mechanical properties that occur following zoledronate treatment. Skeletally mature male C57Bl/6J mice were treated with vehicle (VEH), zoledronate (ZOL), or ZOL followed by raloxifene (RAL; 2 different doses). At the conclusion of 8 weeks of treatment, femora were collected and assessed with microCT and mechanical testing. Trabecular BV/TV was significantly higher in all treated animals compared to VEH with both RAL groups having significantly higher BV/TV compared to ZOL (+21%). All three drug-treated groups had significantly more cortical bone area, higher cortical thickness, and greater moment of inertia at the femoral mid-diaphysis compared to VEH with no difference among the three treated groups. All three drug-treated groups had significantly higher ultimate load compared to VEH-treated animals (+14 to 18%). Both doses of RAL resulted in significantly higher displacement values compared to ZOL-treated animals (+25 to +50%). In conclusion, the current work shows beneficial effects of raloxifene in animals previously treated with zoledronate. The higher mechanical properties of raloxifene-treated animals, combined with similar cortical bone geometry compared to animals treated with zoledronate, suggest that the raloxifene treatment is enhancing mechanical material properties of the tissue.
Collapse
Affiliation(s)
- Cory N Meixner
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, MS 5035, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Mohammad W Aref
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, MS 5035, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Aryaman Gupta
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, MS 5035, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Erin M B McNerny
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, MS 5035, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Drew Brown
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, MS 5035, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Joseph M Wallace
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Engineering, Indiana University Purdue University of Indianapolis, Indianapolis, IN, USA
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, MS 5035, 635 Barnhill Dr., Indianapolis, IN, 46202, USA.
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biomedical Engineering, Indiana University Purdue University of Indianapolis, Indianapolis, IN, USA.
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
20
|
Guss JD, Horsfield MW, Fontenele FF, Sandoval TN, Luna M, Apoorva F, Lima SF, Bicalho RC, Singh A, Ley RE, van der Meulen MC, Goldring SR, Hernandez CJ. Alterations to the Gut Microbiome Impair Bone Strength and Tissue Material Properties. J Bone Miner Res 2017; 32:1343-1353. [PMID: 28244143 PMCID: PMC5466506 DOI: 10.1002/jbmr.3114] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/13/2017] [Accepted: 02/20/2017] [Indexed: 02/07/2023]
Abstract
Alterations in the gut microbiome have been associated with changes in bone mass and microstructure, but the effects of the microbiome on bone biomechanical properties are not known. Here we examined bone strength under two conditions of altered microbiota: (1) an inbred mouse strain known to develop an altered gut microbiome due to deficits in the immune system (the Toll-like receptor 5-deficient mouse [TLR5KO]); and (2) disruption of the gut microbiota (ΔMicrobiota) through chronic treatment with selected antibiotics (ampicillin and neomycin). The bone phenotypes of TLR5KO and WT (C57Bl/6) mice were examined after disruption of the microbiota from 4 weeks to 16 weeks of age as well as without treatment (n = 7 to 16/group, 39 animals total). Femur bending strength was less in ΔMicrobiota mice than in untreated animals and the reduction in strength was not fully explained by differences in bone cross-sectional geometry, implicating impaired bone tissue material properties. Small differences in whole-bone bending strength were observed between WT and TLR5KO mice after accounting for differences in bone morphology. No differences in trabecular bone volume fraction were associated with genotype or disruption of gut microbiota. Treatment altered the gut microbiota by depleting organisms from the phyla Bacteroidetes and enriching for Proteobacteria, as determined from sequencing of fecal 16S rRNA genes. Differences in splenic immune cell populations were also observed; B and T cell populations were depleted in TLR5KO mice and in ΔMicrobiota mice (p < 0.001), suggesting an association between alterations in bone tissue material properties and immune cell populations. We conclude that alterations in the gut microbiota for extended periods during growth may lead to impaired whole-bone mechanical properties in ways that are not explained by bone geometry. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jason D Guss
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Michael W Horsfield
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Fernanda F Fontenele
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Taylor N Sandoval
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Marysol Luna
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Fnu Apoorva
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Svetlana F Lima
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | - Ankur Singh
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Ruth E Ley
- Department of Microbiology, Cornell University, Ithaca, NY, USA
| | - Marjolein Ch van der Meulen
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| | | | - Christopher J Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
21
|
Schlecht SH, Smith LM, Ramcharan MA, Bigelow EM, Nolan BT, Mathis NJ, Cathey A, Manley E, Menon R, McEachin RC, Nadeau JH, Jepsen KJ. Canalization Leads to Similar Whole Bone Mechanical Function at Maturity in Two Inbred Strains of Mice. J Bone Miner Res 2017; 32:1002-1013. [PMID: 28177139 PMCID: PMC5413428 DOI: 10.1002/jbmr.3093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/19/2017] [Accepted: 02/01/2017] [Indexed: 11/10/2022]
Abstract
Previously, we showed that cortical mineralization is coordinately adjusted to mechanically offset external bone size differences between A/J (narrow) and C57BL/6J (wide) mouse femora to achieve whole bone strength equivalence at adulthood. The identity of the genes and their interactions that are responsible for establishing this homeostatic state (ie, canalization) remain unknown. We hypothesize that these inbred strains, whose interindividual differences in bone structure and material properties mimic that observed among humans, achieve functional homeostasis by differentially adjusting key molecular pathways regulating external bone size and mineralization throughout growth. The cortices of A/J and C57BL/6J male mouse femora were phenotyped and gene expression levels were assessed across growth (ie, ages 2, 4, 6, 8, 12, 16 weeks). A difference in total cross-sectional area (p < 0.01) and cortical tissue mineral density were apparent between mouse strains by age 2 weeks and maintained at adulthood (p < 0.01). These phenotypic dissimilarities corresponded to gene expression level differences among key regulatory pathways throughout growth. A/J mice had a 1.55- to 7.65-fold greater expression among genes inhibitory to Wnt pathway induction, whereas genes involved in cortical mineralization were largely upregulated 1.50- to 3.77-fold to compensate for their narrow diaphysis. Additionally, both mouse strains showed an upregulation among Wnt pathway antagonists corresponding to the onset of adult ambulation (ie, increased physiological loads). This contrasts with other studies showing an increase in Wnt pathway activation after functionally isolated, experimental in vivo loading regimens. A/J and C57BL/6J long bones provide a model to develop a systems-based approach to identify individual genes and the gene-gene interactions that contribute to trait differences between the strains while being involved in the process by which these traits are coordinately adjusted to establish similar levels of mechanical function, thus providing insight into the process of canalization. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stephen H Schlecht
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lauren M Smith
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Melissa A Ramcharan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Erin Mr Bigelow
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Bonnie T Nolan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Noah J Mathis
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amber Cathey
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Eugene Manley
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Richard C McEachin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Joseph H Nadeau
- Pacific Northwest Diabetes Research Institute, Seattle, WA, USA
| | - Karl J Jepsen
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
22
|
Ramcharan MA, Faillace ME, Guengerich Z, Williams VA, Jepsen KJ. The development of inter-strain variation in cortical and trabecular traits during growth of the mouse lumbar vertebral body. Osteoporos Int 2017; 28:1133-1143. [PMID: 27734101 PMCID: PMC5890929 DOI: 10.1007/s00198-016-3801-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022]
Abstract
How cortical and trabecular bone co-develop to establish a mechanically functional structure is not well understood. Comparing early postnatal differences in morphology of lumbar vertebral bodies for three inbred mouse strains identified coordinated changes within and between cortical and trabecular traits. These early coordinate changes defined the phenotypic differences among the inbred mouse strains. INTRODUCTION Age-related changes in cortical and trabecular traits have been well studied; however, very little is known about how these bone tissues co-develop from day 1 of postnatal growth to establish functional structures by adulthood. In this study, we aimed to establish how cortical and trabecular tissues within the lumbar vertebral body change during growth for three inbred mouse strains that express wide variation in adult bone structure and function. METHODS Bone traits were quantified for lumbar vertebral bodies of female A/J, C57BL/6J (B6), and C3H/HeJ (C3H) inbred mouse strains from 1 to 105 days of age (n = 6-10 mice/age/strain). RESULTS Inter-strain differences in external bone size were observed as early as 1 day of age. Reciprocal and rapid changes in the trabecular bone volume fraction and alignment in the direction of axial compression were observed by 7 days of age. Importantly, the inter-strain difference in adult trabecular bone volume fraction was established by 7 days of age. Early variation in external bone size and trabecular architecture was followed by progressive increases in cortical area between 28 and 105 days of age, with the greatest increases in cortical area seen in the mouse strain with the lowest trabecular mass. CONCLUSION Establishing the temporal changes in bone morphology for three inbred mouse strains revealed that genetic variation in adult trabecular traits were established early in postnatal development. Early variation in trabecular architecture preceded strain-specific increases in cortical area and changes in cortical thickness. This study established the sequence of how cortical and trabecular traits co-develop during growth, which is important for identifying critical early ages to further focus on intervention studies that optimize adult bone strength.
Collapse
Affiliation(s)
- M A Ramcharan
- Department of Biomedical Engineering, The City College of New York c/o CUNY Graduate Center, New York, NY, USA
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Orthopaedic Surgery, University of Michigan, 109 Zina Pitcher Place, Room 2001, Ann Arbor, MI, 48109-2200, USA
| | - M E Faillace
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Z Guengerich
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - V A Williams
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - K J Jepsen
- Department of Orthopaedic Surgery, University of Michigan, 109 Zina Pitcher Place, Room 2001, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
23
|
Aref MW, McNerny EMB, Brown D, Jepsen KJ, Allen MR. Zoledronate treatment has different effects in mouse strains with contrasting baseline bone mechanical phenotypes. Osteoporos Int 2016; 27:3637-3643. [PMID: 27439372 PMCID: PMC5543625 DOI: 10.1007/s00198-016-3701-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022]
Abstract
UNLABELLED Two strains of mice with distinct bone morphologies and mechanical properties were treated with zoledronate. Our results show a different response to drug treatment in the two strains providing evidence that baseline properties of structure/material may influence response to zoledronate. INTRODUCTION Bisphosphonates are highly effective in reducing fracture risk, yet some individuals treated with these agents still experience fracture. The goal of this study was to test the hypothesis that genotype influences the effect of zoledronate on bone mechanical properties. METHODS Skeletally mature male mice from genetic backgrounds known to have distinct baseline post-yield properties (C57/B6, high post-yield displacement; A/J, low post-yield displacement) were treated for 8 weeks with saline (VEH) or zoledronate (ZOL, 0.06 mg/kg subcutaneously once every 4 weeks) in a 2 × 2 study design. Ex vivo μCT and mechanical testing (4-pt bending) were conducted on the femur to assess morphological and mechanical differences. RESULTS Significant drug and/or genotype effects were found for several mechanical properties and significant drug × genotype interactions were found for measures of strength (ultimate force) and brittleness (total displacement, strain to failure). Treatment with ZOL affected bone biomechanical measures of brittleness (total displacement (-25 %) and strain to failure (-23 %)) in B6 mice significantly differently than in A/J mice. This was driven by unique drug × genotype effects on bone geometry in B6 animals yet likely also reflected changes to the tissue properties. CONCLUSION These data may support the concept that properties of the bone geometry and/or tissue at the time of treatment initiation play a role in determining the bone's mechanical response to zoledronate treatment.
Collapse
Affiliation(s)
- M W Aref
- Department of Anatomy and Cell Biology, MS 5035, Indiana University School of Medicine, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - E M B McNerny
- Department of Anatomy and Cell Biology, MS 5035, Indiana University School of Medicine, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - D Brown
- Department of Anatomy and Cell Biology, MS 5035, Indiana University School of Medicine, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - K J Jepsen
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - M R Allen
- Department of Anatomy and Cell Biology, MS 5035, Indiana University School of Medicine, 635 Barnhill Dr., Indianapolis, IN, 46202, USA.
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
24
|
Ip V, Toth Z, Chibnall J, McBride-Gagyi S. Remnant Woven Bone and Calcified Cartilage in Mouse Bone: Differences between Ages/Sex and Effects on Bone Strength. PLoS One 2016; 11:e0166476. [PMID: 27829059 PMCID: PMC5102366 DOI: 10.1371/journal.pone.0166476] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/28/2016] [Indexed: 11/27/2022] Open
Abstract
Introduction Mouse models are used frequently to study effects of bone diseases and genetic determinates of bone strength. Murine bones have an intracortical band of woven bone that is not present in human bones. This band is not obvious under brightfield imaging and not typically analyzed. Due to the band’s morphology and location it has been theorized to be remnant bone from early in life. Furthermore, lamellar and woven bone are well known to have differing mechanical strengths. The purpose of this study was to determine (i) if the band is from early life and (ii) if the woven bone or calcified cartilage contained within the band affect whole bone strength. Woven Bone Origin Studies In twelve to fourteen week old mice, doxycycline was used to label bone formed prior to 3 weeks old. Doxycycline labeling and woven bone patterns on contralateral femora matched well and encompassed an almost identical cross-sectional area. Also, we highlight for the first time in mice the presence of calcified cartilage exclusively within the band. However, calcified cartilage could not be identified on high resolution cone-beam microCT scans when examined visually or by thresholding methods. Mechanical Strength Studies Subsequently, three-point bending was used to analyze the effects of woven bone and calcified cartilage on whole bone mechanics in a cohort of male and female six and 13 week old Balb/C mice. Three-point bending outcomes were correlated with structural and compositional measures using multivariate linear regression. Woven bone composed a higher percent of young bones than older bones. However, calcified cartilage in older bones was twice that of younger bones, which was similar when normalized by area. Area and/or tissue mineral density accounted for >75% of variation for most strength outcomes. Percent calcified cartilage added significant predictive power to maximal force and bending stress. Calcified cartilage and woven bone could have more influence in genetic models where calcified cartilage percent is double our highest value.
Collapse
Affiliation(s)
- Victoria Ip
- School of Operations Research and Information Engineering, Cornell University, Ithaca, New York, United States of America
| | - Zacharie Toth
- Department of Orthopaedic Surgery, Saint Louis University, Saint Louis, Missouri, United States of America
| | - John Chibnall
- Department of Psychiatry, Saint Louis University, Saint Louis, Missouri, United States of America
| | - Sarah McBride-Gagyi
- Department of Orthopaedic Surgery, Saint Louis University, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
25
|
Ryan JW, Starczak Y, Tsangari H, Sawyer RK, Davey RA, Atkins GJ, Morris HA, Anderson PH. Sex-related differences in the skeletal phenotype of aged vitamin D receptor global knockout mice. J Steroid Biochem Mol Biol 2016; 164:361-368. [PMID: 26690785 DOI: 10.1016/j.jsbmb.2015.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 11/09/2015] [Accepted: 12/06/2015] [Indexed: 10/22/2022]
Abstract
The role of the vitamin D receptor (VDR) in maintaining skeletal health appears to be complex and dependent on the physiological context. Global Vdr deletion in a mouse model (Vdr-/-) results in hypocalcemia, secondary hyperparathyroidism and bone features typical of vitamin D-dependent rickets type II. When weanling Vdr-/- mice are fed a diet containing high levels of calcium, phosphorus and lactose, termed the rescue diet, normalisation of serum calcium, phosphate and parathyroid hormone levels results in prevention of rickets at 10 weeks of age. However, 17 week old male Vdr-/- mice, fed the rescue diet, have been reported as osteopenic due to a decrease in bone formation when compared to wild type mice. We now report confirmation of this finding with further data on the effect of the rescue diet on appendicular and axial skeletal structures in male and female Vdr-/- mice at 26 weeks of age compared to Vdr+/- controls. All Vdr-/- mice were normocalcemic with no evidence of any mineralization defect. However, male Vdr-/- mice exhibited significantly reduced mineral in femoral and vertebral bones when compared to control littermate Vdr+/- mice, consistent with the previously reported data. In contrast, 26-week-old female Vdr-/- mice demonstrated significantly increased femoral trabecular bone volume although there was decreased vertebral trabecular bone volume, similar to males, and femoral cortical bone volume was unchanged. Thus, the Vdr-/- mouse model displays sex- and site-specific differences in skeletal structures with long-term feeding of a rescue diet. Although the global Vdr-/- ablation does not permit the determination of skeletal mechanisms producing these differences, these data confirm skeletal changes even when fed the rescue diet.
Collapse
Affiliation(s)
- Jackson W Ryan
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia
| | - Yolandi Starczak
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia
| | - Helen Tsangari
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia
| | - Rebecca K Sawyer
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia
| | - Rachel A Davey
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia
| | - Gerald J Atkins
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia
| | - Howard A Morris
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia,.
| | - Paul H Anderson
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia; Centre for Orthopaedic and Trauma Research, School of Medicine, University of Adelaide, Adelaide, SA 5001, Australia
| |
Collapse
|
26
|
Liu DM, Zhao L, Liu TT, Jiao PL, Zhao DD, Shih MS, Tao B, Sun LH, Zhao HY, Liu JM. Rictor/mTORC2 loss in osteoblasts impairs bone mass and strength. Bone 2016; 90:50-8. [PMID: 27262777 DOI: 10.1016/j.bone.2016.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 05/25/2016] [Accepted: 05/29/2016] [Indexed: 11/26/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a Ser/Thr kinase conserved through evolution that coordinates extra cellular signals associated with cell growth. Main functions of mTOR present in the form of two complexes, namely mTORC1 and mTORC2, which are distinct in their unique components, raptor and rictor. In the current study, using a Cre/loxp system, we found an anabolic effect of mTORC2 signaling on skeleton. Osteoblast differentiation was reduced, with down-regulation of mTORC2 signaling activity in primary cultures of osteoblasts that did not contain rictor. Mice with a specific deletion of rictor in mature osteoblasts showed a significant reduction in lean mass and bone mineral density by dual energy x-ray absorptiometry analysis. Micro-computed tomography, histomorphometric, and molecular biological analyses revealed a marked impairment of the cortical bone mass and microarchitecture, as well as minor changes in trabecular bone, of the Rictorob(-/-) mice. Cortical bone mass and thickness of the femoral mid-shaft were dramatically reduced, with unusual increases in porosity and marrow area in Rictorob(-/-) mice. Thinner trabeculae were found in the L4 vertebrae with relatively normal structural indices of trabecular numbers and separation. A lower rate of bone turnover was observed, as the consequence of the decreased individual osteoblast activity and bone resorption. Furthermore, these changes were associated with significantly decreased bone biomechanical properties. In conclusion, expression of rictor in osteoblasts is essential for the maintenance of normal bone remodeling and microarchitecture, especially for the maintenance of the cortical bone.
Collapse
Affiliation(s)
- Dong-Mei Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Lin Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China; Department of Endocrine and Metabolic Diseases, ZhongShan Hospital, FuDan University, Shanghai, China
| | - Ting-Ting Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Pei-Lin Jiao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Dian-Dian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Mei-Shu Shih
- PharmaLegacy Laboratories Co., Ltd., Shanghai, China
| | - Bei Tao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Li-Hao Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Hong-Yan Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China.
| |
Collapse
|
27
|
Jindrová A, Tuma J, Sládek V. Impact of Non-Invasively Induced Motor Deficits on Tibial Cortical Properties in Mutant Lurcher Mice. PLoS One 2016; 11:e0158877. [PMID: 27387489 PMCID: PMC4936670 DOI: 10.1371/journal.pone.0158877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/23/2016] [Indexed: 11/18/2022] Open
Abstract
It has been shown that Lurcher mutant mice have significantly altered motor abilities, regarding their motor coordination and muscular strength because of olivorecebellar degeneration. We assessed the response of the cross-sectional geometry and lacuno-canalicular network properties of the tibial mid-diaphyseal cortical bone to motor differences between Lurcher and wild-type (WT) male mice from the B6CBA strain. The first data set used in the cross-sectional geometry analysis consists of 16 mice of 4 months of age and 32 mice of 9 months of age. The second data set used in the lacunar-canalicular network analysis consists of 10 mice of 4 months of age. We compared two cross-sectional geometry and four lacunar-canalicular properties by I-region using the maximum and minimum second moment of area and anatomical orientation as well as H-regions using histological differences within a cross section. We identified inconsistent differences in the studied cross-sectional geometry properties between Lurcher and WT mice. The biggest significant difference between Lurcher and WT mice is found in the number of canaliculi, whereas in the other studied properties are only limited. Lurcher mice exhibit an increased number of canaliculi (p < 0.01) in all studied regions compared with the WT controls. The number of canaliculi is also negatively correlated with the distance from the centroid in the Lurcher and positively correlated in the WT mice. When the Lurcher and WT sample is pooled, the number of canaliculi and lacunar volume is increased in the posterior Imax region, and in addition, midcortical H-region exhibit lower number of canaliculi, lacuna to lacuna distance and increased lacunar volume. Our results indicate, that the importance of precise sample selection within cross sections in future studies is highlighted because of the histological heterogeneity of lacunar-canalicular network properties within the I-region and H-region in the mouse cortical bone.
Collapse
Affiliation(s)
- Alena Jindrová
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jan Tuma
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Laboratory of Neurodegenerative Disorders, Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Vladimír Sládek
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
28
|
Manley E, Perosky JE, Khoury BM, Reddy AB, Kozloff KM, Alford AI. Thrombospondin-2 deficiency in growing mice alters bone collagen ultrastructure and leads to a brittle bone phenotype. J Appl Physiol (1985) 2015; 119:872-81. [PMID: 26272319 DOI: 10.1152/japplphysiol.00340.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/06/2015] [Indexed: 01/15/2023] Open
Abstract
Thrombospondin-2 (TSP2) is a matricellular protein component of the bone extracellular matrix. Long bones of adult TSP2-deficient mice have increased endosteal bone thickness due to expansion of the osteoblast progenitor cell pool, and these cells display deficits in osteoblastic potential. Here, we investigated the effects of TSP2 deficiency on whole bone geometric and mechanical properties in growing 6-wk-old male and female wild-type and TSP2-knockout (KO) mice. Microcomputed tomography and mechanical testing were conducted on femora and L2 vertebrae to assess morphology and whole bone mechanical properties. In a second series of experiments, femoral diaphyses were harvested from wild-type and TSP2-KO mice. Detergent-soluble type I collagen content was determined by Western blot of right femora. Total collagen content was determined by hydroxyproline analysis of left femora. In a third series of experiments, cortical bone was dissected from the anterior and posterior aspects of the femoral middiaphysis and imaged by transmission electron microscopy to visualize collagen fibrils. Microcomputed tomography revealed minimal structural effects of TSP2 deficiency. TSP2 deficiency imparted a brittle phenotype on cortical bone. Femoral tissue mineral density was not affected by TSP2 deficiency. Instead, transmission electron microscopy revealed less intensely stained collagen fibrils with altered morphology in the extracellular matrix assembled by osteoblasts on the anterior surface of TSP2-KO femora. Femoral diaphyseal bone displayed comparable amounts of total collagen, but the TSP2-KO bones had higher levels of detergent-extractable type I collagen. Together, our data suggest that TSP2 is required for optimal collagen fibrillogenesis in bone and thereby contributes to normal skeletal tissue quality.
Collapse
Affiliation(s)
- Eugene Manley
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | | | | | | | | | | |
Collapse
|
29
|
Bortel EL, Duda GN, Mundlos S, Willie BM, Fratzl P, Zaslansky P. Long bone maturation is driven by pore closing: A quantitative tomography investigation of structural formation in young C57BL/6 mice. Acta Biomater 2015; 22:92-102. [PMID: 25829108 DOI: 10.1016/j.actbio.2015.03.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/28/2015] [Accepted: 03/23/2015] [Indexed: 01/14/2023]
Abstract
During mammalian growth, long bones undergo extensive structural reorganization, transforming from primitive shapes in the limb buds into mature bones. Here we shed light on the steps involved in structural formation of the mineralized tissue in midshafts of C57BL/6 femurs, shortly after birth. By combining 3D micrometer-resolution X-ray microtomography with 2D histology, we study the transformation of the tissue from a partially-mineralized scaffold into a compact bone structure. We identify three growth phases that take place during murine long bone maturation: During a patterning phase (I) mineralized struts form a loosely connected foam-like cortical network. During a transitioning phase (II), the extensive non-mineralized tracts vanish, transforming the foam into a fully continuous mass, by 14 days of age. Concomitantly, closed porosity increases to about ∼ 1.4%, and stays at this level, also found in maturity. During a shaping phase (III), the bones gradually attain their characteristic intricate adult form. Architectured mineral depositioning--first in open foamy scaffolds, and later into solid bone material--is presumably a compromise between the mechanical needs of providing support to the body, and the biological requirements of vascularization and extensive nutritional needs in the early stages of bone formation.
Collapse
Affiliation(s)
- Emely L Bortel
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Research Campus Golm, 14476 Potsdam, Germany; Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Georg N Duda
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, Berlin, Ihnestraße 63-73, 14195 Berlin, Germany; Institute of Medical and Human Genetics, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Bettina M Willie
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Peter Fratzl
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Research Campus Golm, 14476 Potsdam, Germany.
| | - Paul Zaslansky
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
30
|
Jepsen KJ, Silva MJ, Vashishth D, Guo XE, van der Meulen MCH. Establishing biomechanical mechanisms in mouse models: practical guidelines for systematically evaluating phenotypic changes in the diaphyses of long bones. J Bone Miner Res 2015; 30:951-66. [PMID: 25917136 PMCID: PMC4794979 DOI: 10.1002/jbmr.2539] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/03/2015] [Accepted: 04/21/2015] [Indexed: 12/30/2022]
Abstract
Mice are widely used in studies of skeletal biology, and assessment of their bones by mechanical testing is a critical step when evaluating the functional effects of an experimental perturbation. For example, a gene knockout may target a pathway important in bone formation and result in a "low bone mass" phenotype. But how well does the skeleton bear functional loads; eg, how much do bones deform during loading and how resistant are bones to fracture? By systematic evaluation of bone morphological, densitometric, and mechanical properties, investigators can establish the "biomechanical mechanisms" whereby an experimental perturbation alters whole-bone mechanical function. The goal of this review is to clarify these biomechanical mechanisms and to make recommendations for systematically evaluating phenotypic changes in mouse bones, with a focus on long-bone diaphyses and cortical bone. Further, minimum reportable standards for testing conditions and outcome variables are suggested that will improve the comparison of data across studies. Basic biomechanical principles are reviewed, followed by a description of the cross-sectional morphological properties that best inform the net cellular effects of a given experimental perturbation and are most relevant to biomechanical function. Although morphology is critical, whole-bone mechanical properties can only be determined accurately by a mechanical test. The functional importance of stiffness, maximum load, postyield displacement, and work-to-fracture are reviewed. Because bone and body size are often strongly related, strategies to adjust whole-bone properties for body mass are detailed. Finally, a comprehensive framework is presented using real data, and several examples from the literature are reviewed to illustrate how to synthesize morphological, tissue-level, and whole-bone mechanical properties of mouse long bones.
Collapse
Affiliation(s)
- Karl J Jepsen
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - X Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Marjolein CH van der Meulen
- Department of Biomedical Engineering and Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
31
|
Wallace IJ, Judex S, Demes B. Effects of load-bearing exercise on skeletal structure and mechanics differ between outbred populations of mice. Bone 2015; 72:1-8. [PMID: 25460574 DOI: 10.1016/j.bone.2014.11.013] [Citation(s) in RCA: 25] [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: 10/19/2014] [Revised: 11/15/2014] [Accepted: 11/17/2014] [Indexed: 11/18/2022]
Abstract
Effects of load-bearing exercise on skeletal structure and mechanical properties can vary between inbred strains of mice. Here, we examine whether such variation also exists at the population level. An experiment was performed with two outbred mouse stocks that have been reproductively isolated for >120 generations (Hsd:ICR, Crl:CD1). Growing females from each stock were either treated with a treadmill-running regimen for 1 month or served as controls. Limb forces were recorded with a force plate and cage activity monitored to verify that they were similar between stocks. After the experiment, femoral cortical and trabecular bone structure were quantified with micro-CT in the mid-diaphysis and distal metaphysis, respectively, and diaphyseal structural strength was determined with mechanical testing. Among Hsd:ICR mice, running led to significant improvements in diaphyseal bone quantity, structural geometry, and mechanical properties, as well as enhanced trabecular morphology. In contrast, among Crl:CD1 mice, the same running regimen had little effect on cortical and trabecular structure and significantly reduced diaphyseal resistance to fracture. In neither stock was body mass, muscle mass, or cage activity level different between runners and controls. Given that most environmental variables were controlled in this study, the differential effects of exercise on Hsd:ICR and Crl:CD1 bones were likely due to genetic differences between stocks. These results suggest that the benefits of loading for bone may vary between human populations (e.g., ethnic groups), in which case exercise programs and technologies designed to promote bone health with mechanical signals may be more advantageous to certain populations than others.
Collapse
Affiliation(s)
- Ian J Wallace
- Department of Anthropology, Stony Brook University, Stony Brook, NY 11794, USA.
| | - Stefan Judex
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Brigitte Demes
- Department of Anatomical Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
32
|
Evaluation of growth patterns and body composition in C57Bl/6J mice using dual energy X-ray absorptiometry. BIOMED RESEARCH INTERNATIONAL 2014; 2014:253067. [PMID: 25110666 PMCID: PMC4119710 DOI: 10.1155/2014/253067] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/29/2014] [Accepted: 06/16/2014] [Indexed: 11/17/2022]
Abstract
The normal growth pattern of female C57BL/6J mice, from 5 to 30 weeks of age, has been investigated in a longitudinal study. Weight, body surface area (BS), and body mass index (BMI) were evaluated in forty mice. Lean mass and fat mass, bone mineral content (BMC), and bone mineral density (BMD) were monitored by dual energy X-ray absorptiometry (DEXA). Weight and BS increased linearly (16.15 ± 0.64-27.64 ± 1.42 g; 51.13 ± 0.74-79.57 ± 2.15 cm(2), P < 0.01), more markedly from 5 to 9 weeks of age (P < 0.001). BMD showed a peak at 17 weeks (0.0548 ± 0.0011 g/cm(2) ∗ m, P < 0.01). Lean mass showed an evident gain at 9 (15.8 ± 0.8 g, P < 0.001) and 25 weeks (20.5 ± 0.3 g, P < 0.01), like fat mass from 13 to 17 weeks (2.0 ± 0.4-3.6 ± 0.7 g, P < 0.01). BMI and lean mass index (LMI) reached the highest value at 21 weeks (3.57 ± 0.02-0.284 ± 0.010 g/cm(2), resp.), like fat mass index (FMI) at 17 weeks (0.057 ± 0.009 g/cm(2)) (P < 0.01). BMI, weight, and BS showed a moderate positive correlation (0.45-0.85) with lean mass from 5 to 21 weeks. Mixed linear models provided a good prediction for lean mass, fat mass, and BMD. This study may represent a baseline reference for a future comparison of wild-type C57BL/6J mice with models of altered growth.
Collapse
|
33
|
Rodríguez-Carballo E, Gámez B, Sedó-Cabezón L, Sánchez-Feutrie M, Zorzano A, Manzanares-Céspedes C, Rosa JL, Ventura F. The p38α MAPK function in osteoprecursors is required for bone formation and bone homeostasis in adult mice. PLoS One 2014; 9:e102032. [PMID: 25007355 PMCID: PMC4090229 DOI: 10.1371/journal.pone.0102032] [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: 12/19/2013] [Accepted: 06/13/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND p38 MAPK activity plays an important role in several steps of the osteoblast lineage progression through activation of osteoblast-specific transcription factors and it is also essential for the acquisition of the osteoblast phenotype in early development. Although reports indicate p38 signalling plays a role in early skeletal development, its specific contributions to adult bone remodelling are still to be clarified. METHODOLOGY/PRINCIPAL FINDINGS We evaluated osteoblast-specific deletion of p38α to determine its significance in early skeletogenesis, as well as for bone homeostasis in adult skeleton. Early p38α deletion resulted in defective intramembranous and endochondral ossification in both calvaria and long bones. Mutant mice showed reduction of trabecular bone volume in distal femurs, associated with low trabecular thickness. In addition, knockout mice also displayed decreased femoral cortical bone volume and thickness. Deletion of p38α did not affect osteoclast function. Yet it impaired osteoblastogenesis and osteoblast maturation and activity through decreased expression of osteoblast-specific transcription factors and their targets. Furthermore, the inducible Cre system allowed us to control the onset of p38α disruption after birth by removal of doxycycline. Deletion of p38α at three or eight weeks postnatally led to significantly lower trabecular and cortical bone volume after 6 or 12 months. CONCLUSIONS Our data demonstrates that, in addition to early skeletogenesis, p38α is essential for osteoblasts to maintain their function in mineralized adult bone, as bone anabolism should be sustained throughout life. Moreover, our data also emphasizes that clinical development of p38 inhibitors should take into account their potential bone effects.
Collapse
Affiliation(s)
- Edgardo Rodríguez-Carballo
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Beatriz Gámez
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Lara Sedó-Cabezón
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Manuela Sánchez-Feutrie
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Cristina Manzanares-Céspedes
- Departament de Patologia i Terapèutica Experimental, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - José Luis Rosa
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
- * E-mail:
| |
Collapse
|
34
|
Lowe DA, Lepori-Bui N, Fomin PV, Sloofman LG, Zhou X, Farach-Carson MC, Wang L, Kirn-Safran CB. Deficiency in perlecan/HSPG2 during bone development enhances osteogenesis and decreases quality of adult bone in mice. Calcif Tissue Int 2014; 95:29-38. [PMID: 24798737 PMCID: PMC4137566 DOI: 10.1007/s00223-014-9859-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 04/10/2014] [Indexed: 12/22/2022]
Abstract
Perlecan/HSPG2 (Pln) is a large heparan sulfate proteoglycan abundant in the extracellular matrix of cartilage and the lacunocanalicular space of adult bones. Although Pln function during cartilage development is critical, evidenced by deficiency disorders including Schwartz-Jampel Syndrome and dyssegmental dysplasia Silverman-Handmaker type, little is known about its function in development of bone shape and quality. The purpose of this study was to understand the contribution of Pln to bone geometric and mechanical properties. We used hypomorph mutant mice that secrete negligible amount of Pln into skeletal tissues and analyzed their adult bone properties using micro-computed tomography and three-point-bending tests. Bone shortening and widening in Pln mutants was observed and could be attributed to loss of growth plate organization and accelerated osteogenesis that was reflected by elevated cortical thickness at older ages. This effect was more pronounced in Pln mutant females, indicating a sex-specific effect of Pln deficiency on bone geometry. Additionally, mutant females, and to a lesser extent mutant males, increased their elastic modulus and bone mineral densities to counteract changes in bone shape, but at the expense of increased brittleness. In summary, Pln deficiency alters cartilage matrix patterning and, as we now show, coordinately influences bone formation and calcification.
Collapse
Affiliation(s)
- Dylan A. Lowe
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Nadia Lepori-Bui
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Peter V. Fomin
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Laura G. Sloofman
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Xiaozhou Zhou
- University of Delaware, Department of Mechanical Engineering, Newark, DE
| | - Mary C. Farach-Carson
- University of Delaware, Department of Biological Sciences, Newark, DE
- Rice University, Department of Biochemistry and Cell Biology, Houston, TX
| | - Liyun Wang
- University of Delaware, Department of Mechanical Engineering, Newark, DE
| | - Catherine B. Kirn-Safran
- University of Delaware, Department of Biological Sciences, Newark, DE
- author to whom correspondence should be addressed: Catherine Kirn-Safran, University of Delaware, Department of Biological Science, 310 Wolf Hall, Newark, DE 19716, Tel: (302) 831-3249, Fax: (302) 831-2281,
| |
Collapse
|
35
|
Luo Q, Leng H, Wang X, Zhou Y, Rong Q. The role of water and mineral-collagen interfacial bonding on microdamage progression in bone. J Orthop Res 2014; 32:217-23. [PMID: 24122969 DOI: 10.1002/jor.22501] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 09/17/2013] [Indexed: 02/04/2023]
Abstract
Microdamage would be accumulated in bone due to high-intensity training or even normal daily activity, which may consequently cause fragility fracture or stress fracture. On the other hand, microdamage formation serves as a toughening mechanism in bone. However, the mechanisms that control microdamage initiation and accumulation in bone are still poorly understood. Our previous finite element model indicated that different interfacial properties between mineral and collagen in bone may lead to distinct patterns of microdamage accumulation. Therefore, the current study was designed to examine such prediction and to investigate the role of water and mineral-collagen interactions on microdamage accumulation in bone. To address these issues, 48 mice femurs were divided randomly into four groups. These groups were dehydrated or treated with perfluorotripropylamine (PFTA) or NaF solution to change water distribution and mineral-collagen interfacial bonding in bone. After three-point bending fatigue tests, the types of microdamage (i.e., linear microcracks or diffuse damage) formed in bone were compared between different groups. The results suggested that (1) bone tissues with strong mineral-collagen interfacial bonding facilitate the formation of linear microcraks, and (2) water has little contribution to the growth of microcracks.
Collapse
Affiliation(s)
- Qing Luo
- College of Engineering, Peking University, Beijing, 100871, China; Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | | | | | | | | |
Collapse
|
36
|
Courtland HW, Kennedy OD, Wu Y, Gao Y, Sun H, Schaffler MB, Yakar S. Low levels of plasma IGF-1 inhibit intracortical bone remodeling during aging. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1691-1703. [PMID: 22976122 PMCID: PMC3776121 DOI: 10.1007/s11357-012-9469-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 08/31/2012] [Indexed: 06/01/2023]
Abstract
Studies linking insulin-like growth factor-1 (IGF-1) to age-related bone loss in humans have been reported but remain only correlative. In this investigation, we characterized the bone phenotype of aged WT C57BL/6J male mice in comparison to that of C57BL/6J mice with reduced serum IGF-1 levels arising from an igfals gene deletion (ALS knockout (ALSKO)). During the aging process, WT mice showed an increase in fat mass and decrease lean mass while ALSKO mice had stable lean and fat mass values. Skeletal analyses of femora from WT mice revealed an expansion of the marrow area and a significant accumulation of intracortical porosity associated with increased intracortical remodeling. In contrast, ALSKO mice showed only small age-related declines in the amount of cortical bone tissue and minimal intracortical porosity, at 2 years of age. Accordingly, mechanical tests of femora from 2-year-old WT mice revealed reduced stiffness and maximal load when compared to bones from ALSKO mice. We show here that lifelong reductions in serum IGF-1 compromise skeletal size in development leading to slender bones; they are also associated with decreased intracortical bone remodeling and preservation of bone strength during aging.
Collapse
Affiliation(s)
- Hayden-William Courtland
- />Division of Endocrinology, Diabetes and Bone Diseases, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Oran D. Kennedy
- />Department of Biomedical Engineering, City College of New York, CUNY, New York, NY 10031 USA
| | - Yingjie Wu
- />Division of Endocrinology, Diabetes and Bone Diseases, Mount Sinai School of Medicine, New York, NY 10029 USA
- />David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, 345 East 24th Street, New York, NY 10010-4086 USA
| | - Ying Gao
- />Division of Endocrinology, Diabetes and Bone Diseases, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Hui Sun
- />Division of Endocrinology, Diabetes and Bone Diseases, Mount Sinai School of Medicine, New York, NY 10029 USA
- />David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, 345 East 24th Street, New York, NY 10010-4086 USA
| | - Mitchell B. Schaffler
- />Department of Biomedical Engineering, City College of New York, CUNY, New York, NY 10031 USA
| | - Shoshana Yakar
- />Division of Endocrinology, Diabetes and Bone Diseases, Mount Sinai School of Medicine, New York, NY 10029 USA
- />David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, 345 East 24th Street, New York, NY 10010-4086 USA
| |
Collapse
|
37
|
Schlecht SH, Jepsen KJ. Functional integration of skeletal traits: an intraskeletal assessment of bone size, mineralization, and volume covariance. Bone 2013; 56:127-38. [PMID: 23721816 PMCID: PMC7368883 DOI: 10.1016/j.bone.2013.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 05/08/2013] [Accepted: 05/10/2013] [Indexed: 11/24/2022]
Abstract
Understanding the functional integration of skeletal traits and how they naturally vary within and across populations will benefit assessments of functional adaptation directed towards interpreting bone stiffness in contemporary and past humans. Moreover, investigating how these traits intraskeletally vary will guide us closer towards predicting fragility from a single skeletal site. Using an osteological collection of 115 young adult male and female African-Americans, we assessed the functional relationship between bone robustness (i.e. total area/length), cortical tissue mineral density (Ct.TMD), and cortical area (Ct.Ar) for the upper and lower limbs. All long bones demonstrated significant trait covariance (p < 0.005) independent of body size, with slender bones having 25-50% less Ct.Ar and 5-8% higher Ct.TMD compared to robust bones. Robustness statistically explained 10.2-28% of Ct.TMD and 26.6-64.6% of Ct.Ar within male and female skeletal elements. This covariance is systemic throughout the skeleton, with either the slender or robust phenotype consistently represented within all long bones for each individual. These findings suggest that each person attains a unique trait set by adulthood that is both predictable by robustness and partially independent of environmental influences. The variation in these functionally integrated traits allows for the maximization of tissue stiffness and minimization of mass so that regardless of which phenotype is present, a given bone is reasonably stiff and strong, and sufficiently adapted to perform routine, habitual loading activities. Covariation intrinsic to functional adaptation suggests that whole bone stiffness depends upon particular sets of traits acquired during growth, presumably through differing levels of cellular activity, resulting in differing tissue morphology and composition. The outcomes of this intraskeletal examination of robustness and its correlates may have significant value in our progression towards improved clinical assessments of bone strength and fragility.
Collapse
Affiliation(s)
- Stephen H Schlecht
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
38
|
Hayward LN, de Bakker CM, Gerstenfeld LC, Grinstaff MW, Morgan EF. Assessment of contrast-enhanced computed tomography for imaging of cartilage during fracture healing. J Orthop Res 2013; 31:567-73. [PMID: 23165442 PMCID: PMC3761062 DOI: 10.1002/jor.22265] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 10/12/2012] [Indexed: 02/04/2023]
Abstract
Assessment of the early stages of fracture healing via X-rays and computed tomography is limited by the low radio-opacity of cartilage. We validated a method of contrast-enhanced computed tomography (CECT) for non-destructive identification of cartilage within a healing fracture callus. Closed, stabilized fractures in femora of C57BL/6 mice were harvested on post-operative day 9.5 and imaged ex vivo with micro-computed tomography (µCT) before and after incubation in a cationic contrast agent that preferentially accumulates in cartilage due to the high concentration of sulfated glycosaminoglycans in the tissue. Co-registration of the pre- and post-incubation images, followed by image subtraction, enabled two- and three-dimensional delineation of mineralized tissue, soft callus, and cartilage. The areas of cartilage and callus identified with CECT were compared to those identified with the gold-standard method of histomorphometry. No difference was found between the areas of cartilage measured by the two methods (p = 0.999). Callus area measured by CECT was smaller than, but strongly predictive of (R(2) = 0.80, p < 0.001), the corresponding histomorphometric measurements. CECT also enabled qualitative identification of mineralized cartilage. These findings indicate that the CECT method provides accurate, quantitative, and non-destructive visualization of the shape and composition of the fracture callus, even during the early stages of repair when little mineralized tissue is present. The non-destructive nature of this method would allow subsequent analyses, such as mechanical testing, to be performed on the callus, thus enabling higher-throughput, comprehensive investigations of bone healing.
Collapse
Affiliation(s)
- Lauren N.M. Hayward
- Department of Biomedical Engineering, Boston University, 44 Cummington Street, Boston, Massachusetts
| | - Chantal M.J. de Bakker
- Department of Biomedical Engineering, Boston University, 44 Cummington Street, Boston, Massachusetts
| | - Louis C. Gerstenfeld
- Department of Orthopedic Surgery, Boston University School of Medicine, 715 Albany Street, Boston, Massachusetts
| | - Mark W. Grinstaff
- Department of Biomedical Engineering, Boston University, 44 Cummington Street, Boston, Massachusetts,Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts
| | - Elise F. Morgan
- Department of Biomedical Engineering, Boston University, 44 Cummington Street, Boston, Massachusetts,Department of Orthopedic Surgery, Boston University School of Medicine, 715 Albany Street, Boston, Massachusetts,Department of Mechanical Engineering, Boston University, 110 Cummington Street, Boston, Massachusetts
| |
Collapse
|
39
|
Blank RD. Insulin, fat, and bone: multiple interactions lead to complex biology. Transl Res 2013; 161. [PMID: 23177795 PMCID: PMC4362712 DOI: 10.1016/j.trsl.2012.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Robert D Blank
- Department of Medicine, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
40
|
Sharir A, Milgram J, Dubnov-Raz G, Zelzer E, Shahar R. A temporary decrease in mineral density in perinatal mouse long bones. Bone 2013; 52:197-205. [PMID: 23044045 DOI: 10.1016/j.bone.2012.09.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 09/27/2012] [Accepted: 09/28/2012] [Indexed: 11/18/2022]
Abstract
Fetal and postnatal bone development in humans is traditionally viewed as a process characterized by progressively increasing mineral density. Yet, a temporary decrease in mineral density has been described in the long bones of infants in the immediate postnatal period. The mechanism that underlies this phenomenon, as well as its causes and consequences, remain unclear. Using daily μCT scans of murine femora and tibiae during perinatal development, we show that a temporary decrease in tissue mineral density (TMD) is evident in mice. By monitoring spatial and temporal structural changes during normal growth and in a mouse strain in which osteoclasts are non-functional (Src-null), we show that endosteal bone resorption is the main cause for the perinatal decrease in TMD. Mechanical testing revealed that this temporary decrease is correlated with reduced stiffness of the bones. We also show, by administration of a progestational agent to pregnant mice, that the decrease in TMD is not the result of parturition itself. This study provides a comprehensive view of perinatal long bone development in mice, and describes the process as well as the consequences of density fluctuation during this period.
Collapse
Affiliation(s)
- A Sharir
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | | | | | | | | |
Collapse
|
41
|
McCoy SY, Falgowski KA, Srinivasan PP, Thompson WR, Selva EM, Kirn-Safran CB. Serum xylosyltransferase 1 level increases during early posttraumatic osteoarthritis in mice with high bone forming potential. Bone 2012; 51:224-31. [PMID: 22146554 PMCID: PMC3319505 DOI: 10.1016/j.bone.2011.11.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 11/06/2011] [Accepted: 11/15/2011] [Indexed: 11/28/2022]
Abstract
Increased proteoglycan (PG) synthesis is essential for the stimulation of cartilage repair processes that take place during the reversible phase of osteoarthritis (OA). In articular cartilage, xylosyltransferase 1 (Xylt1) is the key enzyme that initiates glycosaminoglycan (GAG) chain synthesis by transferring the first sugar residue to the PG core protein. Biological activity of PGs is closely linked to GAG biosynthesis since their polyanionic nature directly contributes to the proper hydration and elastic properties of the cartilage tissue present at the articular interface. The aim of this study was to investigate whether variations in the level of Xylt1 present in serum can be used to predict OA disease progression. The influence of bone forming activity on the systemic release of this enzyme was addressed by experimentally-inducing OA in mice of two different genetic backgrounds that were previously characterized for their distinct bone metabolism: C57BL/6J (B6, high bone remodelers) or C3H/HeJ (C3H, high bone formers). Serum was collected after medial meniscectomy or sham surgeries in young adult mice of these two strains over a period of 3.5months at which point knee histopathology was assessed. A significant increase in serum Xylt1 levels observed shortly after meniscectomy positively correlated with severe cartilage damage evaluated by histological assessment at later time points in mice of the C3H background. In contrast, no temporal regulation of Xylt1 level was found between meniscectomies and control surgeries in B6 mice, which developed OA at a slower rate. Additionally, longitudinal evaluation of the serum levels of other markers of cartilage/bone metabolism (C1,2C, osteocalcin) did not reveal any association with late knee damages. Our results strongly support the idea that serum Xylt1 has a clinical value for monitoring risk of OA progression in young adults with high bone forming potential. Ultimately, the understanding of posttraumatic mechanisms regulating PG synthesis and their modification by GAG will be essential so that interventions that stimulate cartilage regrowth can be undertaken prior to irreversible destruction of the joint tissue. This article is part of a Special Issue entitled "Osteoarthritis".
Collapse
Affiliation(s)
- Sarah Y. McCoy
- University of Delaware, Department of Biological Sciences, Newark, DE
| | | | | | | | - Erica M. Selva
- University of Delaware, Department of Biological Sciences, Newark, DE
| | | |
Collapse
|
42
|
Pathak S, Vachhani SJ, Jepsen KJ, Goldman HM, Kalidindi SR. Assessment of lamellar level properties in mouse bone utilizing a novel spherical nanoindentation data analysis method. J Mech Behav Biomed Mater 2012; 13:102-17. [PMID: 22842281 DOI: 10.1016/j.jmbbm.2012.03.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/23/2012] [Accepted: 03/26/2012] [Indexed: 11/18/2022]
Abstract
In this work, we demonstrate the viability of using our recently developed data analysis procedures for spherical nanoindentation in conjunction with Raman spectroscopy for studying lamellar-level correlations between the local composition and local mechanical properties in mouse bone. Our methodologies allow us to convert the raw load-displacement datasets to much more meaningful indentation stress-strain curves that accurately capture the loading and unloading elastic moduli, the indentation yield points, as well as the post-yield characteristics in the tested samples. Using samples of two different inbred mouse strains, A/J and C57BL/6J (B6), we successfully demonstrate the correlations between the mechanical information obtained from spherical nanoindentation measurements to the local composition measured using Raman spectroscopy. In particular, we observe that a higher mineral-to-matrix ratio correlated well with a higher local modulus and yield strength in all samples. Thus, new bone regions exhibited lower moduli and yield strengths compared to more mature bone. The B6 mice were also found to exhibit lower modulus and yield strength values compared to the more mineralized A/J strain.
Collapse
Affiliation(s)
- Siddhartha Pathak
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, USA.
| | | | | | | | | |
Collapse
|
43
|
Wallace IJ, Tommasini SM, Judex S, Garland T, Demes B. Genetic variations and physical activity as determinants of limb bone morphology: An experimental approach using a mouse model. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2012; 148:24-35. [DOI: 10.1002/ajpa.22028] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 01/03/2012] [Indexed: 01/16/2023]
|
44
|
Kristensen E, Hallgrímsson B, Morck DW, Boyd SK. Microarchitecture, but not bone mechanical properties, is rescued with growth hormone treatment in a mouse model of growth hormone deficiency. Int J Endocrinol 2012; 2012:294965. [PMID: 22505889 PMCID: PMC3312192 DOI: 10.1155/2012/294965] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 01/02/2012] [Indexed: 11/25/2022] Open
Abstract
Growth hormone (GH) deficiency is related to an increased fracture risk although it is not clear if this is due to compromised bone quality or a small bone size. We investigated the relationship between bone macrostructure, microarchitecture and mechanical properties in a GH-deficient (GHD) mouse model undergoing GH treatment commencing at an early (prepubertal) or late (postpubertal) time point. Microcomputed tomography images of the femur and L4 vertebra were obtained to quantify macrostructure and vertebral trabecular microarchitecture, and mechanical properties were determined using finite element analyses. In the GHD animals, bone macrostructure was 25 to 43% smaller as compared to the GH-sufficient (GHS) controls (P < 0.001). GHD animals had 20% and 19% reductions in bone volume ratio (BV/TV) and trabecular thickness (Tb.Th), respectively. Whole bone mechanical properties of the GHD mice were lower at the femur and vertebra (67% and 45% resp.) than the GHS controls (P < 0.001). Both early and late GH treatment partially recovered the bone macrostructure (15 to 32 % smaller than GHS controls) and the whole bone mechanical properties (24 to 43% larger than GHD animals) although there remained a sustained 27-52% net deficit compared to normal mice (P < 0.05). Importantly, early treatment with GH led to a recovery of BV/TV and Tb.Th with a concomitant improvement of trabecular mechanical properties. Therefore, the results suggest that GH treatment should start early, and that measurements of microarchitecture should be considered in the management of GHD.
Collapse
Affiliation(s)
- Erika Kristensen
- Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr NW, Calgary, AB, Canada T2N 1N4
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Benedikt Hallgrímsson
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada T2N 4N1
- Department of Cell Biology and Anatomy, Faculty of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Douglas W. Morck
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada T2N 4N1
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Steven K. Boyd
- Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr NW, Calgary, AB, Canada T2N 1N4
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada T2N 4N1
- *Steven K. Boyd:
| |
Collapse
|
45
|
DONG XNEIL, LENG HUIJIE, RAN QITAO, WANG XIAODU. FINDING OF MICRODAMAGE MORPHOLOGY DIFFERENCES IN MOUSE FEMORAL BONES WITH DISTINCT MINERALIZATION LEVELS. J MECH MED BIOL 2011. [DOI: 10.1142/s0219519410003757] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Microdamage progression in bone is dependent on the ultrastructure of the tissue. Thus, any pathological changes in bone ultrastructure may be reflected in the pattern and capacity of microdamage accumulation. A previous numerical study of microdamage progression in bone using a probabilistic failure analysis approach predicts that the microdamage morphology (either linear microcracks or diffuse damage) is very sensitive to the level of mineralization in bone, which is also implicated in some experimental observations. To examine the prediction, femurs from two strains of mice (C57BL/6J, N = 10 and C3H/HeJ, N = 11) that have distinct mineralization levels were fatigued under four-point bending to create damage in the bone tissues. After testing, the microdamage morphology of the bone samples was examined using bulk-staining technique with basic fuchsin. The results demonstrate that more linear microcracks are observed in femurs of C3H/HeJ (higher mineralization), whereas more diffuse-like damage is found in C57BL/6J femurs (less mineralized). Compared with linear microcracks, the formation of diffuse damage tends to dissipate more energy and help bone to avoid catastrophic failures. Therefore, results from this study may help explain why highly mineralized bone tends to be more brittle. Observations from this study are consistent with the numerical prediction from the previous study, suggesting that mineralization has a significant effect on the microdamage morphology of bone.
Collapse
Affiliation(s)
- X. NEIL DONG
- Department of Mechanical Engineering, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - HUIJIE LENG
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China, 100191, China
| | - QITAO RAN
- Department of Cellular & Structural Biology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX 78245, USA
| | - XIAODU WANG
- Department of Mechanical Engineering, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| |
Collapse
|
46
|
Hayward LNM, de Bakker CMJ, Lusic H, Gerstenfeld LC, Grinstaff MW, Morgan EFI. MRT letter: Contrast-enhanced computed tomographic imaging of soft callus formation in fracture healing. Microsc Res Tech 2011; 75:7-14. [PMID: 22038692 DOI: 10.1002/jemt.21100] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Formation of a cartilaginous soft callus at the site of a bone fracture is a pivotal stage in the healing process. Noninvasive, or even nondestructive, imaging of soft callus formation can be an important tool in experimental and pre-clinical studies of fracture repair. However, the low X-ray attenuation of cartilage renders the soft callus nearly invisible in radiographs. This study utilized a recently developed, cationic, iodinated contrast agent in conjunction with micro-computed tomography to identify cartilage in fracture calluses in the femora of C57BL/6J and C3H/HeJ mice. Fracture calluses were scanned before and after incubation in the contrast agent. The set of pre-incubation images was registered against and then subtracted from the set of post-incubation images, resulting in a three-dimensional map of the locations of cartilage in the callus, as labeled by the contrast agent. This map was then compared to histology from a previous study. The results showed that the locations where the contrast agent collected in relatively high concentrations were similar to those of the cartilage. The contrast agent also identified a significant difference between the two strains of mice in the percentage of the callus occupied by cartilage, indicating that this method of contrast-enhanced computed tomography may be an effective technique for nondestructive, early evaluation of fracture healing.
Collapse
|
47
|
Lynch ME, Main RP, Xu Q, Schmicker TL, Schaffler MB, Wright TM, van der Meulen MCH. Tibial compression is anabolic in the adult mouse skeleton despite reduced responsiveness with aging. Bone 2011; 49:439-46. [PMID: 21642027 PMCID: PMC3235401 DOI: 10.1016/j.bone.2011.05.017] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Revised: 05/17/2011] [Accepted: 05/19/2011] [Indexed: 10/18/2022]
Abstract
The ability of the skeleton to adapt to mechanical stimuli diminishes with age in diaphyseal cortical bone, making bone formation difficult for adults. However, the effect of aging on adaptation in cancellous bone, tissue which is preferentially lost with age, is not well characterized. To develop a model for early post-menopausal women and determine the effect of aging on cancellous bone adaptation in the adult mouse skeleton, in vivo tibial compression was applied to adult (26 week old) osteopenic female mice using loading parameters, peak applied load and peak diaphyseal strain magnitude, that were previously found to be osteogenic in young, growing (10 week old) mice. A Load-Matched group received the same peak applied loads (corresponding to +2100 με at the medial diaphysis of the tibia) and a Strain-Matched group received the same peak diaphyseal strains (+1200 με, requiring half the load) as the young mice. The effects of mechanical loading on bone mass and architecture in adult mice were assessed using micro-computed tomography and in vivo structural stiffness measures. Adaptation occurred only in the Load-Matched group in both the metaphyseal and diaphyseal compartments. Cancellous bone mass increased 54% through trabecular thickening, and cortical area increased 41% through medullary contraction and periosteal expansion. Adult mice were able to respond to an anabolic stimulus and recover bone mass to levels seen in growing mice; however, the adaptive response was reduced relative to that in 10 week old female mice for the same applied load. Using this osteogenic loading protocol, other factors affecting pathological bone loss can be addressed using an adult osteopenic mouse model.
Collapse
Affiliation(s)
- Maureen E Lynch
- Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
BACKGROUND Advances in diagnostic and treatment regimens that aim to reduce fracture incidence will benefit from a better understanding of how bone morphology and tissue quality define whole-bone mechanical properties. QUESTIONS/PURPOSES The goal of this article was to review what is known about the interactions among morphologic and tissue quality traits and how these interactions contribute to bone quality (ie, whole-bone mechanical function). Several questions were addressed. First, how do interactions among morphology and tissue quality traits relate to functional adaptation? Second, what are the emergent patterns of functionally adapted trait sets in long bones? Third, how effective is phenotypic integration at establishing function across a population? Fourth, what are the emergent patterns of functionally adapted trait sets in corticocancellous structures? Fifth, how do functional interactions change with aging? METHODS A literature review was conducted with papers identified primarily through citations listed in reference sections as well as general searches using Google Scholar and PubMed. RESULTS The interactions among adult traits or phenotypic integration are an emergent property of the compensatory mechanisms complex systems used to establish function or homeostasis. Traits are not regulated independently but vary simultaneously (ie, covary) in specific ways to establish function. This covariation results in individuals acquiring unique sets of traits to establish bone quality. CONCLUSIONS AND CLINICAL RELEVANCE Biologic constraints imposed on the skeletal system result in a population showing a pattern of trait sets that is predictable based on external bone size and that can be used to identify individuals with reduced bone quality relative to their bone size and body size.
Collapse
Affiliation(s)
- Karl J Jepsen
- Leni and Peter W. May Department of Orthopaedics, Mount Sinai School of Medicine, Box 1188, One Gustave Levy Place, New York, NY 10029, USA.
| |
Collapse
|
49
|
Nowlan NC, Jepsen KJ, Morgan EF. Smaller, weaker, and less stiff bones evolve from changes in subsistence strategy. Osteoporos Int 2011; 22:1967-80. [PMID: 20857281 DOI: 10.1007/s00198-010-1390-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 08/30/2010] [Indexed: 10/19/2022]
Abstract
UNLABELLED We propose a computational model with which to examine the evolution of bone. Our results indicate that changes in subsistence strategy have influenced the evolution of bone growth and mechanoregulation, and predict that bone size, stiffness, and structural strength may decrease in future generations, bringing increased risk of fracture and prevalence of osteoporosis. INTRODUCTION Archeological data suggest that bone size and strength have decreased over evolution. We hypothesize that changing evolutionary pressures and levels of physical activity, both arising from changes in subsistence strategy, have affected the evolution of bone. We propose a computational model with which to examine the evolution of bone growth and mechanoregulation due to the transitions from hunter-gatherer to agricultural to modern lifestyles. METHODS The evolution of genes governing growth and mechanoregulation in a population of bones is simulated, where each individual is represented by a 2-D bone cross-section. Genetic variability is assumed to modulate growth through mechanoregulatory factors that direct periosteal expansion, endosteal expansion/infilling, and ash content accretion in response to strains incurred during walking. RESULTS The model predicts decreases in cortical area and section modulus (a measure of structural strength) and increases in maximum compressive strain over the course of the simulation, meaning evolution of smaller, less strong, and less stiff bones is predicted for the population average. The model predicts small but continued decreases in size, strength, and stiffness in modern populations, despite the absence of a strong evolutionary advantage to efficient bones during this phase. CONCLUSION In conclusion, our results show that changing loading regimes and evolutionary pressures may have influenced the evolution of bone growth and mechanoregulation, and predict that bone size and strength may continue to decrease in future generations, bringing increased risk of fracture and prevalence of osteoporosis.
Collapse
Affiliation(s)
- N C Nowlan
- Department of Mechanical Engineering, Boston University, 110 Cummington Street, Boston, MA 02215, USA.
| | | | | |
Collapse
|
50
|
Malet A, Bournaud E, Lan A, Mikogami T, Tomé D, Blais A. Bovine lactoferrin improves bone status of ovariectomized mice via immune function modulation. Bone 2011; 48:1028-35. [PMID: 21303707 DOI: 10.1016/j.bone.2011.02.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 01/28/2011] [Accepted: 02/01/2011] [Indexed: 02/02/2023]
Abstract
We have previously shown that bovine lactoferrin (bLF) supplementation can have a beneficial effect on postmenopausal bone loss by modulating bone formation and resorption. A direct effect of bLF on bone metabolism is support by its presence in mice blood. Moreover we know that LF plays a key role in innate immunity and recent studies have shown its ability to modulate adaptive immunity. In particular bLF ingestion prevents recruitment and activation of immune cells at inflammatory sites. We propose that LF through its ability to modulate maturation and differentiation of leucocytes can participate to abolish the deregulation induced by estrogen deficiency on T cells. This study evaluated the effects of bovine lactoferrin on immune function in ovariectomized mice. We investigated whether bLF ingestion could prevent bone loss via modulation of immune function. Three-month-old female C3H mice were either ovariectomized or sham-operated and fed for 1, 2 or 4 months with a control diet (AIN-93M) or the same diet including 10g bLF/kg diet. Bone mineral density was determined using a Lunar Piximus densitometer. The immune parameters were assessed by flow cytometry. In addition, Real-Time PCR was performed to quantify TNFα expression and plasma cytokines were measured at 4 months with Luminex. Ovariectomy induced significant changes on bone parameters and increased recruitment of macrophages, dendritic cells, and B and T cells associated with T lymphocyte activation in bone marrow. Compared to the control diet, ingestion of bLF-enriched diet for 2 months prevented T cell activation and restored dendritic and B cell populations in the bone micro-environment in ovariectomized mice. Furthermore, TNFα expression in bone was decreased by bLF supplementation after 2 and 4 months. Similarly, a decreased plasma level of TNFα was observed concomitantly to an increase of IL-10 level. In conclusion, these experiments suggest that bLF can mediate the prevention of lymphocyte activation and cytokine release in the bone micro-environment. Dietary bLF supplementation could have a beneficial effect on postmenopausal bone loss by modulating immune function.
Collapse
Affiliation(s)
- Arnaud Malet
- AgroParisTech, UMR914 Nutrition Physiology and Ingestive Behavior, Paris, France.
| | | | | | | | | | | |
Collapse
|