Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Front Cell Infect Microbiol ; 13: 1230568, 2023.
Article in English | MEDLINE | ID: mdl-37829606

ABSTRACT

Introduction: Diabetes mellitus (DM) impairs fracture healing and is associated with susceptibility to infection, which further inhibits fracture healing. While intermittent parathyroid hormone (1-34) (iPTH) effectively improves fracture healing, it is unknown whether infection-associated impaired fracture healing can be rescued with PTH (teriparatide). Methods: A chronic diet-induced type 2 diabetic mouse model was used to yield mice with decreased glucose tolerance and increased blood glucose levels compared to lean-fed controls. Methicillin-resistant Staphylococcus aureus (MRSA) was inoculated in a surgical tibia fracture model to simulate infected fracture, after which mice were treated with a combination of antibiotics and adjunctive teriparatide treatment. Fracture healing was assessed by Radiographic Union Scale in Tibial Fractures (RUST), micro-computed tomography (µCT), biomechanical testing, and histology. Results: RUST score was significantly poorer in diabetic mice compared to their lean nondiabetic counterparts. There were concomitant reductions in micro-computed tomography (µCT) parameters of callus architecture including bone volume/total volume, trabecular thickness, and total mineral density in type 2 diabetes mellitus (T2DM) mice. Biomechanicaltesting of fractured femora demonstrated diminished torsional rigidity, stiffness, and toughness to max torque. Adjuvant teriparatide treatment with systemic antibiotic therapy improved numerous parameters of bone microarchitecture bone volume, increased connectivity density, and increased trabecular number in both the lean and T2DM group. Despite the observation that poor fracture healing in T2DM mice was further impaired by MRSA infection, adjuvant iPTH treatment significantly improved fracture healing compared to antibiotic treatment alone in infected T2DM fractures. Discussion: Our results suggest that teriparatide may constitute a viable adjuvant therapeutic agent to improve bony union and bone microarchitecture to prevent the development of septic nonunion under diabetic conditions.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Methicillin-Resistant Staphylococcus aureus , Mice , Animals , Fracture Healing , Teriparatide/therapeutic use , Teriparatide/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , X-Ray Microtomography , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use
2.
J Bone Miner Res ; 37(7): 1352-1365, 2022 07.
Article in English | MEDLINE | ID: mdl-35616626

ABSTRACT

Fracture healing is impaired in the setting of infection, which begets protracted inflammation. The most problematic causative agent of musculoskeletal infection is methicillin-resistant Staphylococcus aureus (MRSA). We hypothesized that modulation of excessive inflammation combined with cell-penetrating antibiotic treatments facilitates fracture healing in a murine MRSA-infected femoral fracture model. Sterile and MRSA-contaminated open transverse femoral osteotomies were induced in 10-week-old male C57BL/6 mice and fixed via intramedullary nailing. In the initial therapeutic cohort, empty, vancomycin (V), rifampin (R), vancomycin-rifampin (VR), or vancomycin-rifampin-trametinib (VRT) hydrogels were applied to the fracture site intraoperatively. Rifampin was included because of its ability to penetrate eukaryotic cells to target intracellular bacteria. Unbiased screening demonstrated ERK activation was upregulated in the setting of MRSA infection. As such, the FDA-approved mitogen-activated protein kinase kinase (MEK)1-pERK1/2 inhibitor trametinib was evaluated as an adjunctive therapeutic agent to selectively mitigate excessive inflammation after infected fracture. Two additional cohorts were created mimicking immediate and delayed postoperative antibiotic administration. Systemic vancomycin or VR was administered for 2 weeks, followed by 2 weeks of VRT hydrogel or oral trametinib therapy. Hematologic, histological, and cytokine analyses were performed using serum and tissue isolates obtained at distinct postoperative intervals. Radiography and micro-computed tomography (µCT) were employed to assess fracture healing. Pro-inflammatory cytokine levels remained elevated in MRSA-infected mice with antibiotic treatment alone, but increasingly normalized with trametinib therapy. Impaired callus formation and malunion were consistently observed in the MRSA-infected groups and was partially salvaged with systemic antibiotic treatment alone. Mice that received VR alongside adjuvant MEK1-pERK1/2 inhibition displayed the greatest restoration of bone and osseous union. A combinatorial approach involving adjuvant cell-penetrating antibiotic treatments alongside mitigation of excessive inflammation enhanced healing of infected fractures. © 2022 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Femoral Fractures , Fracture Healing , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cytokines , Femoral Fractures/drug therapy , Inflammation/drug therapy , Male , Mice , Mice, Inbred C57BL , Rifampin/pharmacology , Rifampin/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Vancomycin/pharmacology , Vancomycin/therapeutic use , X-Ray Microtomography
3.
J Orthop Res ; 39(2): 402-414, 2021 02.
Article in English | MEDLINE | ID: mdl-33336805

ABSTRACT

Infection is a devastating complication following an open fracture. We investigated whether local rifampin-loaded hydrogel can combat infection and improve healing in a murine model of methicillin-resistant Staphylococcus aureus (MRSA) osteomyelitis. A transverse fracture was made at the tibia midshaft of C57BL/6J mice aged 10-12 weeks and stabilized with an intramedullary pin. A total of 1 × 106 colony-forming units (CFU) of MRSA was inoculated. A collagen-based hydrogel containing low-dose (60 µg) and high-dose (300 µg) rifampin was applied before closure. Postoperative treatment response was assessed through bacterial CFU counts from tissue and hardware, tibial radiographs and microcomputed tomography (µCT), immunohistochemistry, and histological analyses. All untreated MRSA-infected fractures progressed to nonunion by 28 days with profuse MRSA colonization. Infected fractures demonstrated decreased soft callus formation on safranin O stain compared to controls. Areas of dense interleukin-1ß stain were associated with poor callus formation. High-dose rifampin hydrogels reduced the average MRSA load in tissue (p < 0.0001) and implants (p = 0.041). Low-dose rifampin hydrogels reduced tissue bacterial load by 50% (p = 0.021). Among sterile models, 88% achieved union compared to 0% of those infected. Mean radiographic union scale in tibia scores improved from 6 to 8.7 with high-dose rifampin hydrogel (p = 0.024) and to 10 with combination local/systemic rifampin therapy (p < 0.0001). µCT demonstrated reactive bone formation in MRSA infection. Histology demonstrated restored fracture healing with bacterial elimination. Rifampin-loaded hydrogels suppressed osteomyelitis, prevented implant colonization, and improved healing. Systemic rifampin was more effective at eliminating infection and improving fracture healing. Further investigation into rifampin-loaded hydrogels is required to correlate these findings with clinical efficacy.


Subject(s)
Antibiotics, Antitubercular/administration & dosage , Fractures, Open/complications , Osteomyelitis/drug therapy , Rifampin/administration & dosage , Staphylococcal Infections/drug therapy , Animals , Bacterial Load/drug effects , Drug Evaluation, Preclinical , Fracture Healing/drug effects , Hydrogels , Male , Methicillin-Resistant Staphylococcus aureus , Mice, Inbred C57BL , Osteomyelitis/etiology , Staphylococcal Infections/etiology
4.
J Nutr Biochem ; 52: 54-61, 2018 02.
Article in English | MEDLINE | ID: mdl-29149648

ABSTRACT

Dried plum (DP), a rich source of polyphenols has been shown to have bone-preserving properties in both animal models of osteoporosis and postmenopausal women. We evaluated if DP alleviated the destruction of joints in transgenic mice (TG) that overexpress human tumor necrosis factor (TNF), a genetic model of rheumatoid arthritis (RA). A four-week treatment of 20% DP diet in TG slowed the onset of arthritis and reduced bone erosions in the joints compared to TG on a regular diet. This was associated with fewer tartrate-resistant acid phosphatase (TRAP) positive cells, suggesting decreased osteoclastogenesis. A DP diet also produced significant protection of articular cartilage and reduction of synovitis. Cultures of human synovial fibroblast in the presence of TNF showed a significant increase in inflammatory interleukin (IL)-1ß, chemokines (monocyte chemoattractant protein-1: MCP1 & macrophage inflammatory protein-1 alpha: MIP1α), cartilage matrix metalloproteinases (MMP1&3), and an osteoclastogenic cytokine (receptor activator of nuclear factor-κB ligand: RANKL) compared to controls. Addition of neochlorogenic acid (NC), a major polyphenol in DP to these cultures resulted in down-regulation of these genes. In the cultures of mouse bone marrow macrophage, NC also repressed TNF-induced formation of osteoclasts and mRNA levels of cathepsin K and MMP9 through inhibition of nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) expression and nuclear factor kappa B (NF-κB) activation. Our data suggested that dietary supplementation with DP inhibited TNF singling; leading to decreased erosions of bone and articular cartilage as well as synovitis.


Subject(s)
Arthritis, Rheumatoid/diet therapy , Chlorogenic Acid/analogs & derivatives , Prunus domestica , Quinic Acid/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Rheumatoid/physiopathology , Bone Resorption/diet therapy , Bone Resorption/drug therapy , Cartilage, Articular/drug effects , Cartilage, Articular/physiopathology , Cell Differentiation/drug effects , Cells, Cultured , Chlorogenic Acid/pharmacology , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Osteogenesis/drug effects , Prunus domestica/chemistry , Quinic Acid/pharmacology , Synoviocytes/drug effects , Synoviocytes/metabolism , Synoviocytes/pathology , Synovitis/diet therapy , Synovitis/prevention & control
5.
J Orthop Res ; 35(12): 2658-2666, 2017 12.
Article in English | MEDLINE | ID: mdl-28460421

ABSTRACT

Osteoclasts play key roles in bone remodeling and pathologic osteolytic disorders such as inflammation, infection, bone implant loosening, rheumatoid arthritis, metastatic bone cancers, and pathological fractures. Osteoclasts are formed by the fusion of monocytes in response to receptor activators of NF-κB-ligand (RANKL) and macrophage colony stimulating factor 1 (M-CSF). Calreticulin (CRT), a commonly known intracellular protein as a calcium-binding chaperone, has an unexpectedly robust anti-osteoclastogenic effect when its recombinant form is applied to osteoclast precursors in vitro or at the site of bone inflammation externally in vivo. Externally applied Calreticulin was internalized inside the cells. It inhibited key pro-osteoclastogenic transcription factors such as c-Fos and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1)-in osteoclast precursor cells that were treated with RANKL in vitro. Recombinant human Calreticulin (rhCRT) inhibited lipopolysaccharide (LPS)-induced inflammatory osteoclastogenesis in the mouse calvarial bone in vivo. Cathepsin K molecular imaging verified decreased Cathepsin K activity when rhCalreticulin was applied at the site of LPS application in vivo. Recombinant forms of intracellular proteins or their derivatives may act as novel extracellular therapeutic agents. We anticipate our findings to be a starting point in unraveling hidden extracellular functions of other intracellular proteins in different cell types of many organs for new therapeutic opportunities. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2658-2666, 2017.


Subject(s)
Bone Resorption/prevention & control , Calreticulin/therapeutic use , Osteogenesis/drug effects , Amino Acid Chloromethyl Ketones , Animals , Calcium Signaling/drug effects , Calreticulin/pharmacology , Cell Line , Drug Evaluation, Preclinical , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , NFATC Transcription Factors/antagonists & inhibitors , Proteomics
6.
J Neurosci ; 32(21): 7137-45, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623658

ABSTRACT

Tau is a microtubule (MT)-stabilizing protein that is altered in Alzheimer's disease (AD) and other tauopathies. It is hypothesized that the hyperphosphorylated, conformationally altered, and multimeric forms of tau lead to a disruption of MT stability; however, direct evidence is lacking in vivo. In this study, an in vivo stable isotope-mass spectrometric technique was used to measure the turnover, or dynamicity, of MTs in brains of living animals. We demonstrated an age-dependent increase in MT dynamics in two different tau transgenic mouse models, 3xTg and rTg4510. MT hyperdynamicity was dependent on tau expression, since a reduction of transgene expression with doxycycline reversed the MT changes. Treatment of rTg4510 mice with the epothilone, BMS-241027, also restored MT dynamics to baseline levels. In addition, MT stabilization with BMS-241027 had beneficial effects on Morris water maze deficits, tau pathology, and neurodegeneration. Interestingly, pathological and functional benefits of BMS-241027 were observed at doses that only partially reversed MT hyperdynamicity. Together, these data suggest that tau-mediated loss of MT stability may contribute to disease progression and that very low doses of BMS-241027 may be useful in the treatment of AD and other tauopathies.


Subject(s)
Cognition Disorders/drug therapy , Epothilones/therapeutic use , Microtubules/pathology , Nerve Degeneration/drug therapy , Tauopathies/drug therapy , Tubulin Modulators/therapeutic use , tau Proteins/physiology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition Disorders/complications , Cognition Disorders/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/psychology , Epothilones/pharmacology , Female , Hippocampus/drug effects , Hippocampus/pathology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/drug effects , Tauopathies/complications , Tauopathies/genetics , Tauopathies/pathology , Tauopathies/psychology , Tubulin Modulators/pharmacology , tau Proteins/antagonists & inhibitors , tau Proteins/biosynthesis , tau Proteins/genetics
7.
J Long Term Eff Med Implants ; 21(2): 159-67, 2011.
Article in English | MEDLINE | ID: mdl-22043974

ABSTRACT

Bone defects resulting from trauma or pathology represent a common and significant clinical problem. In this study, hydroxyapatite (HAp)-alumina bi-layered scaffolds, which have the benefits of both HAp (i.e., osteointegration, osteoconduction) and alumina (i.e., hardness) were used as a bone substitute for the repair of large segmental defects (20 mm) created in a beagle tibia model. Highly porous bi-layered scaffolds with isotropic-pore structures were fabricated using a polymer-template coating technique. The pore sizes obtained using this approach ranged between 230 µm and 470 µm, and porosity was 91.61±1.28%. Using scanning electron microscopy and energy dispersive spectroscopy, it was confirmed that the frame of each bi-layered scaffold consisted of an alumina inner layer and HAp outer layer. The evaluation of bone regeneration within each scaffold after implantation in the beagle tibia was performed using CT, micro-CT, scintigraphy. New bone formation was evident in the large segmental defects treated with HAp/alumina scaffolds. It was concluded from this study that the HAp/alumina bi-layered scaffold is instrumental in inducing host-scaffold engraftment at the distal and proximal ends of the defect as well as distributing the newly formed bone throughout each scaffold 8 weeks post-implantation.


Subject(s)
Aluminum Oxide/pharmacology , Biocompatible Materials/pharmacology , Bone Regeneration , Durapatite/pharmacology , Tibia/injuries , Tibia/physiology , Animals , Dogs , Microscopy, Electron, Scanning , Models, Animal , Pilot Projects , Tibia/ultrastructure , Tissue Scaffolds
8.
Int Orthop ; 34(4): 599-603, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19343345

ABSTRACT

Bone allografts are often hampered by graft incorporation and poor host bone formation. Bisphosphonates, synthetic pyrophosphate analogs, have shown promise in inhibiting bone resorption in human and animal trials. Some in vitro studies have suggested that high dose bisphosphonate may also inhibit bone formation, leading to our hypothesis that an ideal dose of bisphosphonate in allografts could protect allografts from resorption. We transplanted intercalary allografts in to the segmental defect of the rat femurs after soaking each allograft in zoledronate solution (30 microM) and then analysed bone density of the allografts six to 12 weeks after transplantation. At six and 12 weeks, the bone mineral density was higher in the experimental group compared with the control group. Qualitative radiographic and histological analysis also revealed more allograft resorption in the control group than in the zoledronate-treated group. Our data indicate that pharmacological modification of intercalary allografts with zoledronate solution can decrease osteoclast-mediated allograft resorption.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Density/drug effects , Bone Resorption/drug therapy , Diphosphonates/pharmacology , Femur/drug effects , Imidazoles/pharmacology , Animals , Bone Regeneration/drug effects , Bone Remodeling/drug effects , Bone Resorption/diagnostic imaging , Bone Resorption/pathology , Bone Transplantation , Disease Models, Animal , Femur/diagnostic imaging , Femur/metabolism , Graft Survival/drug effects , Graft Survival/physiology , Male , Radiography , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Transplantation, Homologous , Zoledronic Acid
9.
Blood ; 112(4): 1005-12, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18477770

ABSTRACT

Although imatinib, a BCR-ABL tyrosine kinase inhibitor, is used to treat acute Philadelphia chromosome-positive (Ph(+)) leukemia, it does not prevent central nervous system (CNS) relapses resulting from poor drug penetration through the blood-brain barrier. Imatinib and dasa-tinib (a dual-specific SRC/BCR-ABL kinase inhibitor) were compared in a preclinical mouse model of intracranial Ph(+) leukemia. Clinical dasatinib treatment in patients with CNS Ph(+) leukemia was assessed. In preclinical studies, dasatinib increased survival, whereas imatinib failed to inhibit intracranial tumor growth. Stabilization and regression of CNS disease were achieved with continued dasa-tinib administration. The drug also demonstrated substantial activity in 11 adult and pediatric patients with CNS Ph(+) leukemia. Eleven evaluable patients had clinically significant, long-lasting responses, which were complete in 7 patients. In 3 additional patients, isolated CNS relapse occurred during dasatinib therapy; and in 2 of them, it was caused by expansion of a BCR-ABL-mutated dasatinib-resistant clone, implying selection pressure exerted by the compound in the CNS. Dasatinib has promising therapeutic potential in managing intracranial leukemic disease and substantial clinical activity in patients who experience CNS relapse while on imatinib therapy. This study is registered at ClinicalTrials.gov as CA180006 (#NCT00108719) and CA180015 (#NCT00110097).


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System Neoplasms/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics , Adolescent , Adult , Aged , Animals , Child , Cytogenetic Analysis , Dasatinib , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Monitoring , Female , Humans , Male , Mice , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Remission Induction , Spinal Puncture , Survival Rate , Treatment Outcome , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL