Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Anat Rec (Hoboken) ; 305(4): 772-787, 2022 04.
Article in English | MEDLINE | ID: mdl-35226417

ABSTRACT

In 2021, the American Association for Anatomy (AAA) Board of Directors appointed a Task Force on Structural Racism to understand how the laws, rules, and practices in which the Association formed, developed and continues to exist affect membership and participation. This commentary is the first public report from the Task Force. We focus on African Americans with some comments on Jews and women, noting that all marginalized groups deserve study. Through much of its 130 year history, some members were an essential part of perpetuating racist ideas, the Association largely ignored racism and had some practices that prevented participation. The Task Force concluded that individual and structural racism within the AAA, combined with the broader social context in which the Association developed, contributed to the current underrepresentation of African Americans who constitute 4.1% of the membership even though 13.4% of the U.S. population is Black. Intentional efforts within the AAA to reckon with racism and other forms of bias have only begun in the last 10-20 years. These actions have led to more diverse leadership within the Association, and it is hoped that these changes will positively affect the recruitment and retention of marginalized people to science in general and anatomy in particular. The Task Force recommends that the AAA Board issue a statement of responsibility to acknowledge its history. Furthermore, the Task Force advocates that the Board commit to (a) sustaining ongoing projects to improve diversity, equity, and inclusion and (b) dedicating additional resources to facilitate novel initiatives.


Subject(s)
Racism , Black or African American , Female , Humans , Systemic Racism , United States
2.
J Orthop Res ; 40(4): 862-870, 2022 04.
Article in English | MEDLINE | ID: mdl-34061392

ABSTRACT

Bone microarchitectural parameters significantly contribute to implant fixation strength but the role of bone matrix composition is not well understood. To determine the relative contribution of microarchitecture and bone matrix composition to implant fixation strength, we placed titanium implants in 12-week-old intact Sprague-Dawley rats, ovariectomized-Sprague-Dawley rats, and Zucker diabetic fatty rats. We assessed bone microarchitecture by microcomputed tomography, bone matrix composition by Raman spectroscopy, and implant fixation strength at 2, 6, and 10 weeks postimplantation. A stepwise linear regression model accounted for 83.3% of the variance in implant fixation strength with osteointegration volume/total volume (50.4%), peri-implant trabecular bone volume fraction (14.2%), cortical thickness (9.3%), peri-implant trabecular crystallinity (6.7%), and cortical area (2.8%) as the independent variables. Group comparisons indicated that osseointegration volume/total volume was significantly reduced in the ovariectomy group at Week 2 (~28%) and Week 10 (~21%) as well as in the diabetic group at Week 10 (~34%) as compared with the age matched Sprague-Dawley group. The crystallinity of the trabecular bone was significantly elevated in the ovariectomy group at Week 2 (~4%) but decreased in the diabetic group at Week 10 (~3%) with respect to the Sprague-Dawley group. Our study is the first to show that bone microarchitecture explains most of the variance in implant fixation strength, but that matrix composition is also a contributing factor. Therefore, treatment strategies aimed at improving bone-implant contact and peri-implant bone volume without compromising matrix quality should be prioritized.


Subject(s)
Implants, Experimental , Osseointegration , Animals , Female , Humans , Ovariectomy , Rats , Rats, Sprague-Dawley , Rats, Zucker , Titanium , X-Ray Microtomography/methods
3.
FASEB J ; 34(11): 14302-14317, 2020 11.
Article in English | MEDLINE | ID: mdl-32931052

ABSTRACT

Particles released from implants cause inflammatory bone loss, which is a key factor in aseptic loosening, the most common reason for joint replacement failure. With the anticipated increased incidence of total joint replacement in the next decade, implant failure will continue to burden patients. The gut microbiome is increasingly recognized as an important factor in bone physiology, however, its role in implant loosening is currently unknown. We tested the hypothesis that implant loosening is associated with changes in the gut microbiota in a preclinical model. When the particle challenge caused local joint inflammation, decreased peri-implant bone volume, and decreased implant fixation, the gut microbiota was affected. When the particle challenge did not cause this triad of local effects, the gut microbiota was not affected. Our results suggest that cross-talk between these compartments is a previously unrecognized mechanism of failure following total joint replacement.


Subject(s)
Gastrointestinal Microbiome , Inflammation/pathology , Osteolysis/pathology , Prostheses and Implants/adverse effects , Prosthesis-Related Infections/pathology , Animals , Inflammation/etiology , Male , Osteolysis/etiology , Prosthesis-Related Infections/etiology , Rats
4.
JBMR Plus ; 4(4): e10344, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32258964

ABSTRACT

To test how osteoporosis drugs affect bone matrix maturation during cortical bone remodeling, 72 pregnant rats were switched from a 0.4% to a 0.01% calcium diet at parturition for a 23-day lactation period. At weaning, eight dams were sacrificed to establish baseline values, while the remaining dams were returned to 0.4% calcium and treated with vehicle (saline), sodium fluoride (NaF), zoledronic acid (ZA), or sclerostin antibody (Scl-Ab) for either 7 or 28 days (eight animals per group per time point). Femora were examined by µCT, dynamic histomorphometry, Fourier transform infrared imaging, and three-point bending of notched specimens. Cortical porosity decreased in all groups from baseline to day 28. Intracortical mineralizing surface (MS/BS) and mineral apposition rate (MAR), as well as the mineral-to-matrix ratio were unaffected by treatment, but intracortical crystallinity was increased in the ZA group at day 10 compared with vehicle. Cortical area increased in all groups over 28 days mainly because of an addition of bone at the endocortical surface. Endocortical MS/BS did not vary among the groups, but endocortical MAR was suppressed in the NaF group at day 2 and elevated in the Scl-Ab group at day 4 compared with vehicle. Endocortical mineral-to-matrix ratio was increased at days 5 and 10 following NaF treatment and endocortical crystallinity was increased at day 5 following ZA treatment compared with vehicle. Fracture toughness did not differ among the groups. Thus, the treatments affected matrix maturation more strongly at the endocortical then intracortical envelope. In this model of induced remodeling, the bone formation phase is synchronized at multiple sites, facilitating study of the effects of drugs or other bone-targeting agents on matrix maturation independent of their effects on the initiation of remodeling. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

5.
J Orthop Res ; 38(6): 1208-1215, 2020 06.
Article in English | MEDLINE | ID: mdl-31821588

ABSTRACT

Mechanical fixation of the implant to host bone is an important contributor to orthopedic implant survivorship. The relative importance of bone-implant contact, trabecular bone architecture, and cortical bone geometry to implant fixation strength has never been directly tested, especially in the settings of differential implant surface properties. Thus, using a rat model where titanium rods were placed into the intramedullary canal of the distal femur, we determined the relative contribution of bone-implant contact and peri-implant bone architecture to the fixation strength in implants with different surface roughness: highly polished and smooth (as-received) and dual acid-etched (DAE) implants. Using a training set that maximized variance in implant fixation strength, we initially examined correlation between implant fixation strength and outcome parameters from microcomputed tomography and found that osseointegration volume per total volume (OV/TV), trabecular bone volume per total volume (BV/TV), and cortical thickness (Ct.Th) were the single best compartment-specific predictors of fixation strength. We defined separate regression models to predict implant fixation strength for as-received and DAE implants. When the training set models were applied to independent validation sets, we found strong correlations between predicted and experimentally measured implant fixation strength, with r2 = .843 in as received and r2 = .825 in DAE implants. Interestingly, for as-received implants, OV/TV explained more of the total variance in implant fixation strength than the other variables, whereas in DAE implants, Ct.Th had the most explanatory power, suggesting that surface topography of implants affects which bone compartment is most important in providing implant fixation strength.


Subject(s)
Implants, Experimental , Animals , Biomechanical Phenomena , Calcification, Physiologic , Osseointegration , Rats , Rats, Sprague-Dawley , Surface Properties
6.
J Orthop Res ; 38(6): 1216-1227, 2020 06.
Article in English | MEDLINE | ID: mdl-31825107

ABSTRACT

Biomarkers are of interest to identify patients at risk for peri-implant osteolysis and aseptic loosening. We used a rat model of particle-induced peri-implant osteolysis to investigate if early changes in biomarkers were associated with subsequent implant fixation strength. Implants were placed in rat femora, which were then challenged with intra-articular knee injections of either clean polyethylene, lipopolysaccharide-doped polyethylene, or cobalt-chromium alloy particles, with particle-free vehicle serving as control (n ≥ 8 per group). Rats were weighed weekly, blood was collected at weeks 0, 3, 5, and 6, and locomotor behavior was assessed 4 days before study conclusion. Rats were euthanized 6 weeks post surgery. Week 6 serum was analyzed for five bone remodeling markers, while longitudinal serum was assessed for osteocalcin. Bone-implant contact, peri-implant trabecular architecture, and implant fixation strength were measured. Rats challenged with cobalt-chromium particles had a significant reduction in implant fixation strength compared with the vehicle-control group (P = .034). This group also had elevated serum osteocalcin (P = .005), depressed weight gain (P = .001) and less frequent rearing behavior (P = .029). Regardless of group, change in serum osteocalcin at week 3 (r = -.368; P = .046), change in weight at week 2 (r = .586; P < .001), as well as weight change at all other time intervals were associated with fixation strength. The finding that early alterations in serum osteocalcin and body weight were predictive of subsequent implant fixation strength supports continued investigation of biomarkers for early detection of peri-implant osteolysis and implant loosening. Further, change in biomarker levels was found to be more indicative of implant fixation status than any single measurement.


Subject(s)
Body Weight , Implants, Experimental/adverse effects , Osteocalcin/blood , Animals , Biomarkers/blood , Bone Remodeling , Lipopolysaccharides/pharmacology , Male , Motor Activity , Osteolysis , Polyethylene/pharmacology , Rats , Rats, Sprague-Dawley
7.
J Orthop Res ; 36(10): 2754-2761, 2018 10.
Article in English | MEDLINE | ID: mdl-29873110

ABSTRACT

Peri-implant osteolysis is commonly diagnosed after substantial bone loss has occurred, making revision surgery more challenging. The goal of the current study was to identify urinary biomarkers that differentiate total hip replacement patients who eventually develop osteolysis from patients who do not. We used a repository of 24-h urine samples collected prior to surgery and annually thereafter in 26 patients, 16 who developed osteolysis, and 10 who did not. We examined the markers at radiographic diagnosis, annually for 6 years preceding diagnosis, at the first post-operative sampling point, and pre-operatively. Patients in the osteolysis and non-osteolysis groups were matched according to time post-surgery and did not differ in the male:female ratio or age at surgery. Seven candidate biomarkers were measured, including free deoxypyridinoline (DPD), cross-linked N-telopeptides (NTX), interleukin-6 (IL-6), interleukin-8 (IL-8), osteoprotegerin (OPG), α-crosslaps (α-CTX), and ß-crosslaps (ß-CTX). As an individual biomarker, DPD demonstrated the highest ability to predict osteolysis, with an area under the curve (AUC) in Receiver Operating Characteristic (ROC) analyses of 0.844 at 6 years prior to diagnosis. A panel of α-CTX and IL-6 was able to identify at-risk patients with an AUC of 0.941 or greater at all post-operative time points and an AUC of 1.000 pre-operatively. The results demonstrate the potential of using non-invasive biomarkers to identify patients at risk for peri-implant osteolysis long before the emergence of radiographic signs. Further, the high accuracy of the pre-operative biomarker levels demonstrates the potential importance of pre-existing, patient-specific factors driving subsequent osteolysis. Study Design © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2754-2761, 2018.


Subject(s)
Arthroplasty, Replacement, Hip/adverse effects , Biomarkers/urine , Osteolysis/urine , Aged , Female , Humans , Male , Middle Aged , Osteolysis/diagnostic imaging , Osteolysis/etiology , Radiography
8.
J Orthop Res ; 36(3): 979-986, 2018 03.
Article in English | MEDLINE | ID: mdl-28851105

ABSTRACT

Histology and backscatter scanning electron microscopy (bSEM) are the current gold standard methods for quantifying bone-implant contact (BIC), but are inherently destructive. Microcomputed tomography (µCT) is a non-destructive alternative, but attempts to validate µCT-based assessment of BIC in animal models have produced conflicting results. We previously showed in a rat model using a 1.5 mm diameter titanium implant that the extent of the metal-induced artefact precluded accurate measurement of bone sufficiently close to the interface to assess BIC. Recently introduced commercial laboratory µCT scanners have smaller voxels and improved imaging capabilities, possibly overcoming this limitation. The goals of the present study were to establish an approach for optimizing µCT imaging parameters and to validate µCT-based assessment of BIC. In an empirical parametric study using a 1.5 mm diameter titanium implant, we determined 90 kVp, 88 µA, 1.5 µm isotropic voxel size, 1600 projections/180°, and 750 ms integration time to be optimal. Using specimens from an in vivo rat experiment, we found significant correlations between bSEM and µCT for BIC with the manufacturer's automated analysis routine (r = 0.716, p = 0.003) or a line-intercept method (r = 0.797, p = 0.010). Thus, this newer generation scanner's improved imaging capability reduced the extent of the metal-induced artefact zone enough to permit assessment of BIC. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:979-986, 2018.


Subject(s)
Bone and Bones/diagnostic imaging , Prostheses and Implants , X-Ray Microtomography/methods , Animals , Titanium
9.
J Orthop Res ; 35(12): 2740-2748, 2017 12.
Article in English | MEDLINE | ID: mdl-28471533

ABSTRACT

Articular cartilage lines the load-bearing surfaces of long bones and undergoes compositional and structural degeneration during osteoarthritis progression. Contrast enhanced microcomputed tomography (µCT) is being applied to a variety of preclinical models, including the mouse, to map structural and compositional properties in 3-D. The thinness (∼30-50 µm) and high cellularity of mouse articular cartilage presents a significant imaging challenge. Our group previously showed that mouse articular cartilage and proteoglycan (PG) content can be assessed by µCT with the ioxagalate-based contrast agent Hexabrix, but the voxel size used (6 µm) was deemed to be barely adequate. The objective of the present study is to assess the utility of a novel contrast agent, CA4+, to quantify mouse articular cartilage morphology and composition with high resolution µCT imaging (3 µm voxels) and to compare the sensitivity of CA4+ and Hexabrix to detect between-group differences. While both contrast agents are iodine-based, Hexabrix is anionic and CA4+ is cationic so they interact differently with negatively charged PGs. With CA4+, a strong correlation was found between non-calcified articular cartilage thickness measurements made with histology and µCT (R2 = 0.72, p < 0.001). Cartilage degeneration-as assessed by loss in volume, thickness, and PG content-was observed in 34-week-old mice when compared to both 7- and 12-week-old mice. High measurement precision was observed with CA4+, with the coefficient of variation after repositioning and re-imaging samples equaling 2.8%, 4.5%, 7.4% and 5.9% for attenuation, thickness, volume, and PG content, respectively. Use of CA4+ allowed increased sensitivity for assessing PG content compared to Hexabrix, but had no advantage for measurement of cartilage thickness or volume. This improvement in imaging should prove useful in preclinical studies of cartilage degeneration and regeneration. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2740-2748, 2017.


Subject(s)
Cartilage, Articular/diagnostic imaging , Contrast Media , Animals , Drug Evaluation, Preclinical , Female , Ioxaglic Acid , Mice, Inbred C57BL , Trypsin , X-Ray Microtomography
10.
J Orthop Res ; 33(9): 1374-81, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25808034

ABSTRACT

Various intact and post-injury bone phenotypes are heritable traits. In this study, we sought to determine if intramembranous bone regeneration following marrow ablation differed among common inbred mouse strains and to identify how early the differences appear. We found a ∼four-fold difference in the regenerated bone volume 21 days after marrow ablation in females from four inbred mouse strains: FVB/N (15.7 ± 8.1%, mean and standard deviation), C3H/He (15.5 ± 4.2%), C57BL/6 (12.2 ± 5.2%), and BALB/c (4.0 ± 4.4%); with BALB/c different from FVB/N (p = 0.007) and C3H/He (p = 0.002). A second experiment showed that FVB/N compared to BALB/c mice had more regenerated bone 7 and 14 days after ablation (p < 0.001), while at 21 days FVB/N mice had a greater fraction of mineralizing surface (p = 0.008) without a difference in mineral apposition rate. Thus, differences among strains are evident early during intramembranous bone regeneration following marrow ablation and appear to be associated with differences in osteogenic cell recruitment, but not osteoblast activity. The amount of regenerating bone was not correlated with other heritable traits such as the intact bone phenotype or soft tissue wound healing, suggesting that there may be independent genetic pathways for these traits.


Subject(s)
Bone Marrow/pathology , Bone Regeneration , Animals , Bone and Bones/pathology , Female , Femur/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Osteoblasts/cytology , Osteogenesis/physiology , Phenotype , Regeneration , Species Specificity
11.
J Bone Joint Surg Am ; 97(2): 133-40, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25609440

ABSTRACT

BACKGROUND: The mechanical fixation of orthopaedic and dental implants is compromised by diminished bone volume, such as with osteoporosis. Systemic administration of sclerostin antibody (Scl-Ab) has been shown to enhance implant fixation in normal animals. In the present study, we tested whether Scl-Ab can improve implant fixation in established osteoporosis in a rat model. METHODS: We used an ovariectomized (ovx) rat model, in which we found a 78% decrease in trabecular bone volume at the time of implant surgery; sham-ovx, age-matched rats were used as controls. After placement of a titanium implant in the medullary cavity of the distal aspect of the femur, the rats were maintained for four, eight, or twelve weeks and treated biweekly with Scl-Ab or with the delivery vehicle alone. Outcomes were measured with use of microcomputed tomography, mechanical testing, and static and dynamic histomorphometry. RESULTS: Scl-Ab treatment doubled implant fixation strength in both the sham-ovx and ovx groups, although the enhancement was delayed in the ovx group. Scl-Ab treatment also enhanced bone-implant contact; increased peri-implant trabecular thickness and volume; and increased cortical thickness. These structural changes were associated with an approximately five to sevenfold increase in the bone-formation rate and a >50% depression in the eroded surface following Scl-Ab treatment. Trabecular bone thickness and bone-implant contact accounted for two-thirds of the variance in fixation strength. CONCLUSIONS: In this model of severe osteoporosis, Scl-Ab treatment enhanced implant fixation by stimulating bone formation and suppressing bone resorption, leading to enhanced bone-implant contact and improved trabecular bone volume and architecture. CLINICAL RELEVANCE: Systemic administration of anti-sclerostin antibodies might be a useful strategy in total joint replacement when bone mass is deficient.


Subject(s)
Bone Morphogenetic Proteins/administration & dosage , Osseointegration/drug effects , Osteoporosis/complications , Prosthesis Failure/drug effects , Animals , Antibodies/administration & dosage , Bone Morphogenetic Proteins/immunology , Bone Resorption/etiology , Bone Resorption/prevention & control , Disease Models, Animal , Female , Genetic Markers/immunology , Osteogenesis/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley
12.
J Orthop Surg Res ; 9: 91, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25288055

ABSTRACT

BACKGROUND: Metal-on-metal prostheses undergo wear and corrosion, releasing soluble ions and wear particles into the surrounding environment. Reports described early failures of the metal-on-metal prostheses, with histologic features similar to a Type IV immune response. Mechanisms by which metal wear products and metal ion causing this reaction are not completely understood, and the effects of metal ions on osteocytes, which represent more than 95% of all the bone cells, have not been also studied. We hypothesized that soluble metal ions released from the cobalt-chromium-molybdenum (Co-Cr-Mo) prosthesis may have cytotoxic effect on osteocytes. METHODS: MLO-Y4 osteocytes were treated with various metal ion solutions for 24 and 48 h. The effect of ion treatment on cytotoxicity was assessed by WST-1 reagents and cell death ELISA. Morphological changes were analyzed by a phase-contrast microscope or fluorescent microscope using Hoechst 33342 and propidium iodine staining. RESULTS: Cr and Mo ions did not cause cell death under 0.50 mM, highest concentration studied, whereas Co and Ni ions had significant cytotoxic effect on MLO-Y4 cells at concentrations grater than 0.10 mM and at 0.50 mM, respectively, in a dose-dependent manner. According to the ELISA data, osteocytes treated with Co ions were more susceptible to necrotic than apoptotic cell death, while Ni ions caused osteocyte apoptosis. The morphological assays show that cells treated with Co and Ni ions at high concentration were fewer in number and rounded. In addition, fluorescent images showed a marked reduction in live cells and an increase in dead osteocytes treated with Co and Ni ions at high concentration. CONCLUSIONS: Metal ions released from metal-on-metal bearing surfaces have potentially cytotoxic effects on MLO-Y4 osteocytes, in vitro.


Subject(s)
Cobalt/toxicity , Nickel/toxicity , Osteocytes/drug effects , Animals , Cell Death/drug effects , Cell Line , Chromium/toxicity , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Transgenic , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Molybdenum/toxicity
13.
J Orthop Trauma ; 28(10): 605-11, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24464096

ABSTRACT

BACKGROUND: Bone repair is regulated by biological factors and the local mechanical environment. We hypothesize that the combined use of low-intensity pulsed ultrasound (LIPUS) and recombinant human bone morphogenetic protein-2 (rhBMP-2) will synergistically or additively enhance bone regeneration in a model simulating the more difficult scenarios in orthopaedic traumatology. METHODS: Femoral defects in rats were replaced with absorbable collagen sponges carrying rhBMP-2 (0, 1.2, 6, or 12 µg; n = 30). Each group was divided equally to receive daily treatment of either LIPUS or sham stimulation. At 4 weeks, new bone formation was assessed using quantitative (radiography and microcomputed tomography), qualitative (histology), and functional (biomechanical) end points. RESULTS: LIPUS with 1.2 µg of rhBMP-2 significantly improved the radiographic healing as compared with its sham control starting as early as 2 weeks. Quantitatively, the use of LIPUS with 6 µg of rhBMP-2 significantly increased the bone volume. However, using LIPUS with 12 µg of rhBMP-2 indicated a reduction in callus size, without compromising the bone volume, which was also observable histologically, showing organized lamellar bone and repopulated marrow in the original defect region. Histologically, 1.2 µg of rhBMP-2 alone showed the presence of uncalcified cartilage in the defect, which was reduced with LIPUS treatment. Biomechanically, LIPUS treatment significantly increased the peak torsion and stiffness in the 6- and 12 µg rhBMP-2 groups. CONCLUSIONS: LIPUS enhances rhBMP-2-induced bone formation at lower doses (1.2 and 6 µg) and callus maturation at 12-µg dose delivered on absorbable collagen sponge for bone repair in a rat critical-sized femoral segmental defect.


Subject(s)
Bone Morphogenetic Protein 2/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Osteogenesis/drug effects , Osteogenesis/radiation effects , Transforming Growth Factor beta/administration & dosage , Ultrasonic Therapy , Animals , Bone Regeneration/drug effects , Bone Regeneration/physiology , Bone Regeneration/radiation effects , Disease Models, Animal , Femur/drug effects , Femur/physiopathology , Femur/radiation effects , Male , Osteogenesis/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage
14.
J Orthop Res ; 31(5): 800-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23280449

ABSTRACT

To examine bone remodeling following implant placement, 88 female Sprague-Dawley rats underwent either sham ovariectomy (sham-ovx) or ovariectomy (ovx) at 4.5 months. At 11 months, 17 baseline control animals were euthanized, while 71 rats received bilateral intramedullary femoral implants. Implanted rats were randomized to 4-, 8-, or 12-week follow-up times. Microcomputed tomography was used to assess cortical area and trabecular architecture in all rats. Dynamic and static histomorphometry were performed in a subset to examine the trabecular and endocortical bone in the distal femoral metaphysis adjacent to the implant and the periosteal surface at the midshaft superior to the implant (n = 59). Implantation did not affect bone volume in either sham-ovx or ovx rats compared to baseline controls. Implant placement significantly increased mineralizing surface, mineral apposition rate, and bone formation rate in both sham-ovx and ovx rats at the trabecular and endocortical surfaces at four and sometimes 8 weeks, with a return to baseline values by 12 weeks. At the periosteal surface, implant placement increased bone formation at 4 weeks with a return to baseline levels by 8 weeks. Thus, implant placement increases bone remodeling transiently without affecting bone volume in sham-ovx and ovx rats.


Subject(s)
Bone Remodeling/physiology , Osseointegration/physiology , Osteoporosis/physiopathology , Osteoporosis/surgery , Prostheses and Implants , Animals , Disease Models, Animal , Female , Femur/diagnostic imaging , Femur/physiopathology , Femur/surgery , Materials Testing , Osteoporosis/diagnostic imaging , Ovariectomy , Periosteum/diagnostic imaging , Periosteum/physiopathology , Periosteum/surgery , Radiography , Random Allocation , Rats , Rats, Sprague-Dawley
15.
Arthritis Rheum ; 64(12): 4012-20, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23192793

ABSTRACT

OBJECTIVE: To assess the ability of sclerostin antibody therapy to blunt the negative effects of polyethylene particles on implant fixation and peri-implant bone structure in a rat implant fixation model. METHODS: Thirty-six adult male rats received intramedullary titanium implants; 12 rats received vehicle injections only (control), and 24 rats received intraarticular injections of lipopolysaccharide-doped polyethylene particles. Twelve of the rats that received particles also received sclerostin antibody treatment. The 3 groups of rats were maintained for 12 weeks in a pathogen-free environment, at which time mechanical, micro-computed tomography, and dynamic and static histomorphometry end points were assessed. RESULTS: Sclerostin antibody treatment completely blocked the negative effect of the lipopolysaccharide-doped polyethylene particles on implant fixation and peri-implant bone volume by increasing the bone formation rate and depressing bone resorption. CONCLUSION: Anabolic agents targeting the Wnt signaling pathway are a promising new alternative for the prevention of periprosthetic osteolysis and aseptic loosening.


Subject(s)
Antibodies/pharmacology , Bone Morphogenetic Proteins/immunology , Bone Resorption/prevention & control , Genetic Markers/immunology , Osteogenesis/drug effects , Polyethylene/adverse effects , Prostheses and Implants , Prosthesis Failure/etiology , Animals , Antibodies/immunology , Antibodies/therapeutic use , Bone Resorption/physiopathology , Femur/diagnostic imaging , Femur/physiology , Femur/surgery , Male , Models, Animal , Osteogenesis/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Titanium , Tomography, X-Ray Computed , Wnt Proteins/physiology
16.
J Tissue Eng ; 3(1): 2041731412459745, 2012.
Article in English | MEDLINE | ID: mdl-22962632

ABSTRACT

An exogenous supply of growth factors and bioreplaceable scaffolds may help bone regeneration. The aim of this study was to examine the effects of TGF-ß1 and VEGF-A transgenes on the osteogenic potential of bone marrow stromal cells. Rat bone marrow stromal cells were transfected with plasmids encoding mouse TGF-ß1 and/or VEGF-A complementary DNAs and cultured for up to 28 days. Furthermore, collagen scaffolds carrying combinations of the plasmids-transfected cells were implanted subcutaneously in rats. The transgenes increased alkaline phosphatase activity, enhanced mineralized nodule formation, and elevated osteogenic gene expressions in vitro. In vivo, messenger RNA expression of osteogenic genes such as BMPs and Runx2 elevated higher by the transgenes. The data indicate that exogenous TGF-ß1 and VEGF-A acted synergistically and could induce osteoblastic differentiation of bone marrow stromal cells in both cell culture and an animal model. The results may provide valuable information to optimize protocols for transgene-and-cell-based tissue engineering.

17.
J Bone Joint Surg Am ; 94(18): 1670-80, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22992878

ABSTRACT

BACKGROUND: Previous studies have demonstrated that sclerostin blockade is anabolic for bone. This study examined whether systemic administration of sclerostin antibody would increase implant fixation and peri-implant bone volume in a rat model. METHODS: Titanium cylinders were placed in the femoral medullary canal of ninety male Sprague-Dawley rats. One-half of the rats (n=45) received murine sclerostin antibody (Scl-Ab, 25 mg/kg, twice weekly) and the other one-half (n=45) received saline solution. Equal numbers of rats from both groups were sacrificed at two, four, or eight weeks after the implant surgery and the femora were examined by microcomputed tomography, mechanical pull-out testing, and histology. RESULTS: Fixation strength in the two groups was similar at two weeks but was 1.9-fold greater at four weeks (p=0.024) and 2.2-fold greater at eight weeks (p<0.001) in the rats treated with sclerostin antibody. At two weeks, antibody treatment led to increased cortical area, with later increases in cortical thickness and total cross-sectional area. Significant differences in peri-implant trabecular bone were not evident until eight weeks but included increased bone volume per total volume, bone structure that was more plate-like, and increased trabecular thickness and number. Changes in bone architecture in the intact contralateral femur tended to precede the peri-implant changes. The peri-implant bone properties accounted for 61% of the variance in implant fixation strength, 32% of the variance in stiffness, and 63% of the variance in energy to failure. The implant fixation strength at four weeks was approximately equivalent to the strength in the control group at eight weeks. CONCLUSIONS: Sclerostin antibody treatment accelerated and enhanced mechanical fixation of medullary implants in a rat model by increasing both cortical and trabecular bone volume.


Subject(s)
Antibodies, Monoclonal/pharmacology , Femur/diagnostic imaging , Femur/surgery , Osteogenesis/drug effects , Prosthesis Implantation/methods , Animals , Biomechanical Phenomena , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Risk Assessment , Tensile Strength , Titanium , Treatment Outcome , X-Ray Microtomography/methods
18.
Clin Orthop Relat Res ; 470(9): 2503-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22528386

ABSTRACT

BACKGROUND: Stem cell mobilization, which is defined as the forced egress of stem cells from the bone marrow to the peripheral blood (PB) using chemokine receptor agonists, is an emerging concept for enhancing tissue regeneration. However, the effect of stem cell mobilization by a single injection of the C-X-C chemokine receptor type 4 (CXCR4) antagonist AMD3100 on intramembranous bone regeneration is unclear. QUESTIONS/PURPOSES: We therefore asked: Does AMD3100 mobilize adult stem cells in C57BL/6 mice? Are stem cells mobilized to the PB after marrow ablation? And does AMD3100 enhance bone regeneration? METHODS: Female C57BL/6 mice underwent femoral marrow ablation surgery alone (n = 25), systemic injection of AMD3100 alone (n = 15), or surgery plus AMD3100 (n = 57). We used colony-forming unit assays, flow cytometry, and micro-CT to investigate mobilization of mesenchymal stem cells, endothelial progenitor cells, and hematopoietic stem cells to the PB and bone regeneration. RESULTS: AMD3100 induced mobilization of stem cells to the PB, resulting in a 40-fold increase in mesenchymal stem cells. The marrow ablation injury mobilized all three cell types to the PB over time. Administration of AMD3100 led to a 60% increase in bone regeneration at Day 21. CONCLUSIONS: A single injection of a CXCR4 antagonist lead to stem cell mobilization and enhanced bone volume in the mouse marrow ablation model of intramembranous bone regeneration.


Subject(s)
Adult Stem Cells/drug effects , Bone Regeneration/drug effects , Cell Movement/drug effects , Femur/drug effects , Hematopoietic Stem Cell Mobilization/methods , Heterocyclic Compounds/pharmacology , Adult Stem Cells/immunology , Animals , Benzylamines , Bone Marrow/drug effects , Bone Marrow/surgery , Cell Proliferation/drug effects , Colony-Forming Units Assay , Cyclams , Endothelial Cells/drug effects , Female , Femur/diagnostic imaging , Femur/immunology , Femur/surgery , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Models, Animal , Pilot Projects , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Regeneration/drug effects , Time Factors , X-Ray Microtomography
19.
J Biomed Mater Res A ; 100(4): 918-28, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22275163

ABSTRACT

Revision surgery for particle-induced implant loosening in total joint replacement is expected to increase dramatically over the next few decades. This study was designed to investigate if local tissue and serum markers of bone remodeling reflect implant fixation following administration of lipopolysaccharide (LPS)-doped polyethylene (PE) particles in a rat model. Twenty-four rats received bilateral implantation of intramedullary titanium rods in the distal femur, followed by weekly bilateral intra-articular injection of either LPS-doped PE particles (n = 12) or vehicle that contained no particles (n = 12) for 12 weeks. The group in which the particles were injected had increased serum C-terminal telopeptide of type I collagen (CTX-I), decreased serum osteocalcin (OC), increased peri-implant eroded surface, decreased peri-implant bone volume, and decreased mechanical pull-out strength compared to the controls. Implant fixation strength was positively correlated with peri-implant bone volume and serum OC and inversely correlated with serum CTX-I, while energy to yield was positively correlated with serum OC and inversely correlated with the number of tartrate-resistant acid phosphatase positive cells at the interface and the amount of peri-implant eroded surface. There was no effect on trabecular bone volume at a remote site. Thus, the particle-induced impaired fixation in this rat model was directly associated with local and serum markers of elevated bone resorption and depressed bone formation, supporting the rationale of exploring both anticatabolic and anabolic strategies to treat and prevent particle-related implant osteolysis and loosening, and indicating that serum markers may prove useful in tracking implant fixation.


Subject(s)
Biomarkers , Bone Remodeling , Prostheses and Implants , Animals , Male , Models, Animal , Rats , Rats, Sprague-Dawley
20.
J Mech Behav Biomed Mater ; 5(1): 1-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22100074

ABSTRACT

Dentin matrix protein-1 (DMP1) is a key regulator of biomineralization. Here, we examine changes in structural, geometric, and material properties of cortical bone in a transgenic mouse model overexpressing DMP1. Micro-computed tomography and three-point bending were performed on 90 femora of wild type and transgenic mice at 1, 2, 4, and 6 months. Fourier transform infrared imaging was performed at 2 months. We found that the transgenic femurs were longer (p<0.01), more robust in cross-section (p<0.05), stronger (p<0.05), but had less post-yield strain and displacement (p<0.01), and higher tissue mineral density (p<0.01) than the wild type femurs at 1 and 2 months. At 2 months, the transgenic femurs also had a higher mineral-to-matrix ratio (p<0.05) and lower carbonate substitution (p<0.05) compared to wild type femurs. These findings indicate that increased mineralization caused by overexpressing DMP1 led to increased structural cortical bone properties associated with decreased ductility during the early post-natal period.


Subject(s)
Calcification, Physiologic/genetics , Extracellular Matrix Proteins/genetics , Femur/metabolism , Mechanical Phenomena , Animals , Biomechanical Phenomena , Femur/diagnostic imaging , Femur/physiology , Gene Expression , Materials Testing , Mice , Mice, Transgenic , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...