Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
JBMR Plus ; 7(11): e10819, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38025036

ABSTRACT

An increasing number of patients with type 2 diabetes (T2DM) will require total joint replacement (TJR) in the next decade. T2DM patients are at increased risk for TJR failure, but the mechanisms are not well understood. The current study used the Zucker Diabetic-Sprague Dawley (ZDSD) rat model of T2DM with Sprague Dawley (SPD) controls to investigate the effects of intramedullary implant placement on osseointegration, peri-implant bone structure and matrix composition, and fixation strength at 2 and 10 weeks post-implant placement. Postoperative inflammation was assessed with circulating MCP-1 and IL-10 2 days post-implant placement. In addition to comparing the two groups, stepwise linear regression modeling was performed to determine the relative contribution of glucose, cytokines, bone formation, bone structure, and bone matrix composition on osseointegration and implant fixation strength. ZDSD rats had decreased peri-implant bone formation and reduced trabecular bone volume per total volume compared with SPD controls. The osseointegrated bone matrix of ZDSD rats had decreased mineral-to-matrix and increased crystallinity compared with SPD controls. Osseointegrated bone volume per total volume was not different between the groups, whereas implant fixation was significantly decreased in ZDSD at 2 weeks but not at 10 weeks. A combination of trabecular mineral apposition rate and postoperative MCP-1 levels explained 55.6% of the variance in osseointegration, whereas cortical thickness, osseointegration mineral apposition rate, and matrix compositional parameters explained 69.2% of the variance in implant fixation strength. The results support the growing recognition that both peri-implant structure and matrix composition affect implant fixation and suggest that postoperative inflammation may contribute to poor outcomes after TJR surgeries in T2DM patients. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

2.
Acad Med ; 97(9): 1331-1334, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35263304

ABSTRACT

PROBLEM: Early grant support for junior faculty members appears to positively influence their career trajectory. The authors sought to determine whether provision of grant support that enables early-career faculty members to conduct clinical, basic science, or educational research improves their academic success and enhances retention. APPROACH: The authors compared career development and retention among 30 Cohn Fellowship recipients and 31 nonrecipients who participated in the same mentoring program. An award of $20,000 to the fellowship recipients ensured protected time for research for 1 year. Academic productivity of both groups was monitored for 6 years. OUTCOMES: The authors found statistically significant differences between the 2 groups regarding research funding and scholarly productivity. The Cohn Fellowship recipients received a total of $14.7 million in external funding vs $3.7 million for nonrecipients, reflecting mean funding of $588,116 and $196,658 per person, respectively ( P < .01). Recipients published a total of 174 peer-reviewed articles vs 26 for nonrecipients, reflecting a mean of 7 and 1 per person, respectively ( P < .01). Recipients gave a total of 268 presentations vs 25 for nonrecipients, with a mean of 11 and 1 per person, respectively ( P < .01). Furthermore, 8 of the 25 recipients who stayed at Rush University (32%) were promoted to associate professor compared with 2 of the 19 (11%) nonrecipients ( P = .15). A majority of the Cohn Fellows (25; 83%) stayed at Rush University during the study compared with 61% of nonrecipients ( P = .06). These findings suggest that even small amounts of research support received early in a career can benefit the faculty and the university as a whole. NEXT STEPS: We plan to continue gathering data to increase sample size and analyze outcomes for specific variables (e.g., time, rank, gender, promotion, retention).


Subject(s)
Awards and Prizes , Mentoring , Faculty, Medical , Fellowships and Scholarships , Financing, Organized , Humans
3.
JBMR Plus ; 6(3): e10591, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35309863

ABSTRACT

Many lifestyle factors affect bone. Sleep deprivation increases risk for fractures and alcohol consumption can lead to alterations in the skeleton. How combined exposure to these two risk factors affects bone is unclear. Thus, we sought to determine the effects of circadian rhythm disruption and chronic alcohol intake on bone structure and mechanical properties in mice. A total of 120 male C57BL/6J mice were used in two cohorts of 60 mice each because of limited availability of light-tight housing cabinets. One cohort was born in winter and the other in summer. Mice were randomly assigned to circadian disruption (weekly shifting of the light/dark cycle) and control (no shifting) groups beginning at 8 to 12 weeks of age for 12 weeks at which time mice were administered an alcohol-containing or control diet for an additional 10 weeks. Bone structure and mechanical properties of the femur were assessed by micro-computed tomography and three-point bending, respectively. The initial data analysis revealed a likely cohort effect. Thus, we used a three-way analysis of variance to assess the effects of circadian rhythm disruption, alcohol intake, and cohort. Circadian rhythm disruption alone had minimal effects on bone structure and mechanical properties. Alcohol intake reduced body mass and had minimal effects on cortical bone regardless of circadian disruption. Alcohol intake resulted in higher trabecular bone volume, but these beneficial effects were blunted when circadian rhythm was disrupted. Cohort significantly affected body size, many cortical bone structure outcomes, some trabecular bone structure outcomes, and tissue-level material properties. Thus, cohort had the predominant effect on bone structure and mechanical properties in this study, with chronic alcohol intake and environmental circadian disruption having less consistent effects. The data indicate that season of birth may affect skeletal phenotypes and that studies requiring multiple cohorts should determine if a cohort effect exists. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

4.
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
5.
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
6.
Curr Osteoporos Rep ; 18(5): 577-586, 2020 10.
Article in English | MEDLINE | ID: mdl-32734511

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to critically evaluate the current literature regarding implant fixation in osteoporotic bone. RECENT FINDINGS: Clinical studies have not only demonstrated the growing prevalence of osteoporosis in patients undergoing total joint replacement (TJR) but may also indicate a significant gap in screening and treatment of this comorbidity. Osteoporosis negatively impacts bone in multiple ways beyond the mere loss of bone mass, including compromising skeletal regenerative capacity, architectural deterioration, and bone matrix quality, all of which could diminish implant fixation. Recent findings both in preclinical animal models and in clinical studies indicate encouraging results for the use of osteoporosis drugs to promote implant fixation. Implant fixation in osteoporotic bone presents an increasing clinical challenge that may be benefitted by increased screening and usage of osteoporosis drugs.


Subject(s)
Bone-Implant Interface , Joint Prosthesis , Osteoporosis/physiopathology , Prosthesis Retention , Animals , Arthroplasty, Replacement , Biomechanical Phenomena , Humans , Prosthesis Failure
7.
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
8.
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
9.
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
10.
J Orthop Res ; 35(12): 2595-2605, 2017 12.
Article in English | MEDLINE | ID: mdl-28548682

ABSTRACT

We completed a systematic literature review of in vivo animal models that use arthrotomy-based methods to study particle-induced peri-implant osteolysis. The purpose of the review was to characterize the models developed to date, to determine the questions addressed, to assess scientific rigor and transparency, and to identify gaps in knowledge. We probed three literature databases (Medline, Embase, and Scopus) and found 77 manuscripts that fit the search parameters. In the most recent 10 years, researchers mainly used rat and mouse models, whereas in the previous 20 years, large animal, canine, and rabbit models were more common. The studies have demonstrated several pathophysiology pathways, including macrophage migration, particle phagocytosis, increased local production of cytokines and lysosomal enzymes, elevated bone resorption, and suppressed bone formation. The effect of variation in particle characteristics and concentration received limited attention with somewhat mixed findings. Particle contamination by endotoxin was shown to exacerbate peri-implant osteolysis. The possibility of early diagnosis was demonstrated through imaging and biomarker approaches. Several studies showed that both local and systemic delivery of bisphosphonates inhibits the development of particle-induced osteolysis. Other methods of inhibiting osteolysis include the use of anabolic agents and altering the implant design. Few studies examined non-surgical rescue of loosened implants, with conflicting results with alendronate. We found that the manuscripts often lacked the methodological detail now advocated by the ARRIVE guidelines, suggesting that improvement in reporting would be useful to maximize rigor and transparency. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2595-2605, 2017.


Subject(s)
Disease Models, Animal , Osteolysis , Animals
11.
Bonekey Rep ; 5: 837, 2016.
Article in English | MEDLINE | ID: mdl-27648259

ABSTRACT

In this paper, we provide a detailed protocol for a model of long bone mechanical marrow ablation in the rodent, including surgical procedure, anesthesia, and pre- and post-operative care. In addition, frequently used experimental end points are briefly discussed. This model was developed to study intramembranous bone regeneration following surgical disruption of the marrow contents of long bones. In this model, the timing of the appearance of bone formation and remodeling is well-characterized and therefore the model is well-suited to evaluate the in vivo effects of various agents which influence these processes. When biomaterials such as tissue engineering scaffolds or metal implants are placed in the medullary cavity after marrow ablation, end points relevant to tissue engineering and implant fixation can also be analyzed. By sharing a detailed protocol, we hope to improve inter-laboratory reproducibility.

12.
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
13.
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
14.
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
15.
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
16.
J Bone Miner Res ; 29(7): 1597-607, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24470143

ABSTRACT

Sclerostin antibody (Scl-Ab) is a novel bone-forming agent that is currently undergoing preclinical and clinical testing. Scl-Ab treatment is known to dramatically increase bone mass, but little is known about the quality of the bone formed during treatment. In the current study, global mineralization of bone matrix in rats and nonhuman primates treated with vehicle or Scl-Ab was assayed by backscattered scanning electron microscopy (bSEM) to quantify the bone mineral density distribution (BMDD). Additionally, fluorochrome labeling allowed tissue age-specific measurements to be made in the primate model with Fourier-transform infrared microspectroscopy to determine the kinetics of mineralization, carbonate substitution, crystallinity, and collagen cross-linking. Despite up to 54% increases in the bone volume after Scl-Ab treatment, the mean global mineralization of trabecular and cortical bone was unaffected in both animal models investigated. However, there were two subtle changes in the BMDD after Scl-Ab treatment in the primate trabecular bone, including an increase in the number of pixels with a low mineralization value (Z5) and a decrease in the standard deviation of the distribution. Tissue age-specific measurements in the primate model showed that Scl-Ab treatment did not affect the mineral-to-matrix ratio, crystallinity, or collagen cross-linking in the endocortical, intracortical, or trabecular compartments. Scl-Ab treatment was associated with a nonsignificant trend toward accelerated mineralization intracortically and a nearly 10% increase in carbonate substitution for tissue older than 2 weeks in the trabecular compartment (p < 0.001). These findings suggest that Scl-Ab treatment does not negatively impact bone matrix quality.


Subject(s)
Antibodies/pharmacology , Bone Matrix/metabolism , Glycoproteins/immunology , Animals , Bone Density/drug effects , Bone Matrix/drug effects , Diaphyses/diagnostic imaging , Diaphyses/drug effects , Diaphyses/ultrastructure , Femur/diagnostic imaging , Femur/drug effects , Femur/ultrastructure , Macaca fascicularis , Male , Microscopy, Fluorescence , Models, Animal , Organ Specificity/drug effects , Rats, Sprague-Dawley , X-Ray Microtomography
17.
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
18.
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
19.
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.

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...