Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Orthop ; 43(6): e427-e432, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-36977364

RESUMEN

BACKGROUND: The globally acknowledged treatment for mild to moderate slipped capital femoral epiphysis (SCFE) is single screw in situ fixation, also used for prophylactic contralateral fixation. The Free-Gliding Screw (FG; Pega Medical) is a 2-part free-extending screw system designed to allow the growth of the proximal femur. We aimed to analyze the relationship between skeletal maturity and potential growth of the proximal physis and remodeling of the femoral neck using this implant. MATERIALS AND METHODS: Females below 12 years and males below 14 years undergoing in situ fixation for stable SCFE or prophylactic fixation were treated using the implant. Three elements of the modified Oxford Bone (mOB 3 ) score were used to measure maturity (triradiate cartilage, head of the femur, and greater trochanter). Radiographs were analyzed immediately postoperatively and at a minimum of 2 years for a change in screw length, posterior-sloping angle, articulotrochanteric distance, α angle, and head-neck offset. RESULTS: The study group comprised 30 (F:M=12:18) of 39 hips treated with SCFE and 22 (F:M=13:9) of 29 hips managed prophylactically using the free-Gliding screw. In the therapeutic group, chronologic age was a less valuable predictor of future screw lengthening than mOB 3 . An mOB 3 of ≤13 predicted future growth of >6 mm but did not reach statistical significance ( P =0.07). Patients with open triradiates showed a mean screw lengthening of 6.6 mm compared with those with closed triradiates (4.0 mm), but this did not reach significance ( P =0.12). In those with mOB 3 ≤13, the α angle reduced significantly ( P <0.01) and the head-neck offset increased significantly, suggesting remodeling. There was no change in these parameters when mOB 3 ≥14. In the prophylactic group, change in screw length was significant with mOB 3 of ≤13 (mean=8.0 mm, P <0.05), as was the presence of an open triradiate cartilage (mean=7.7 mm, P <0.05). In both cohorts, posterior-sloping angle and articulotrochanteric distance did not change, indicating no slip progression in either treatment or prophylactic groups and minimal effect on the proximal physeal growth relative to the greater trochanter. CONCLUSIONS: Growing screw constructs can halt slip progression while allowing proximal femoral growth in young patients with SCFE. Ongoing growth is better when the implant is used for prophylactic fixation. The results in treated SCFE need to be expanded to demonstrate a clinically meaningful cut-off for significant growth, but SCFE patients with an open triradiate remodel significantly more than those where it is closed. LEVEL OF EVIDENCE: Level III-retrospective comparative study.


Asunto(s)
Epífisis Desprendida de Cabeza Femoral , Masculino , Femenino , Humanos , Epífisis Desprendida de Cabeza Femoral/diagnóstico por imagen , Epífisis Desprendida de Cabeza Femoral/cirugía , Estudios Retrospectivos , Fémur/diagnóstico por imagen , Fémur/cirugía , Cuello Femoral/diagnóstico por imagen , Cuello Femoral/cirugía , Placa de Crecimiento
2.
Am J Med Genet A ; 185(10): 2976-2985, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34155781

RESUMEN

Reduced muscle tone, muscle weakness, and physical fatigue can impact considerably on quality of life for children with neurofibromatosis type 1 (NF1). Human muscle biopsies and mouse models of NF1 deficiency in muscle show intramyocellular lipid accumulation, and preclinical data have indicated that L-carnitine supplementation can ameliorate this phenotype. The aim of this study is to examine whether daily L-carnitine supplementation is safe and feasible, and will improve muscle strength and reduce fatigue in children with NF1. A 12-week Phase 2a trial was conducted using 1000 mg daily oral levocarnitine tartrate supplementation. Recruited children were between 8 and 12 years old with a clinical diagnosis of NF1, history of muscle weakness and fatigue, and naïve to L-carnitine. Primary outcomes were safety (self-reporting, biochemical testing) and compliance. Secondary outcomes included plasma acylcarnitine profiles, functional measures (muscle strength, long jump, handwriting speed, 6-minute-walk test [6MWT]), and parent-reported questionnaires (PedsQL™, CBCL/6-18). Six children completed the trial with no self-reported adverse events. Biochemical tests for kidney and liver function were normal, and the average compliance was 95%. Plasma acylcarnitine levels were low, but within a range not clinically linked to carnitine deficiency. For strength measures, there was a mean 53% increase in dorsiflexion strength (95% confidence interval [CI] 8.89-60.75; p = 0.02) and mean 66% increase in plantarflexion strength (95% CI 12.99-134.1; p = 0.03). In terms of muscle performance, there was a mean 10% increase in long jump distance (95% CI 2.97-16.03; p = 0.01) and 6MWT distance (95% CI 5.88-75.45; p = 0.03). Comparison with the 1000 Norms Project data showed a significant improvement in Z-score for all of these measures. Parent reports showed no negative impact on quality of life, and the perceived benefits led to the majority of individuals remaining on L-carnitine after the study. Twelve weeks of L-carnitine supplementation is safe and feasible in children with NF1, and a Phase 3 trial should confirm the efficacy of treatment.


Asunto(s)
Carnitina/administración & dosificación , Fatiga/dietoterapia , Debilidad Muscular/dietoterapia , Neurofibromatosis 1/dietoterapia , Cardiomiopatías/dietoterapia , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Carnitina/efectos adversos , Carnitina/deficiencia , Carnitina/metabolismo , Niño , Suplementos Dietéticos/efectos adversos , Fatiga/genética , Fatiga/patología , Femenino , Humanos , Hiperamonemia/dietoterapia , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Fuerza Muscular/efectos de los fármacos , Debilidad Muscular/metabolismo , Debilidad Muscular/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Enfermedades Musculares/dietoterapia , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/patología , Calidad de Vida
3.
J Paediatr Child Health ; 57(1): 41-45, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32772467

RESUMEN

AIM: Australia utilises a selective ultrasound screening programme. The rate of late diagnosis of developmental dysplasia of the hip (DDH) in Australia is increasing. The aim of this study is to quantify the treatment required and compare the 5-year radiological outcomes between early and late diagnosis in children with DDH with frank dislocation. METHODS: We performed a case-matched control study of children with frank DDH dislocations from 2000 to 2010 comparing three groups: children with an early diagnosis successfully treated in a Pavlik harness (SP), children with an early diagnosis who failed Pavlik harness treatment (FP) and children with a late diagnosis (LD). Minimum follow-up was 4 years. RESULTS: A total of 115 hips were included. Patients in the LD group required significantly more open reductions (P < 0.001), acetabular osteotomies (P < 0.001) and femoral osteotomies (P < 0.001). LD was also associated with significantly higher rates of growth disturbance at 46.3%, compared to 20.6% in the FP group and 5% in the SP group (P < 0.001). Overall, there were excellent radiological outcomes in 58.5% of the LD group compared to 79.4% in the FP group and 100% in the SP group. CONCLUSION: In Australia, high rates of LD in DDH persist in the context of selective ultrasound screening. While good radiological outcomes are achievable, a significantly greater level of surgical intervention is required and this is associated with significantly higher rates of growth disturbance. Optimisation of screening in Australia is vital.


Asunto(s)
Displasia del Desarrollo de la Cadera , Luxación Congénita de la Cadera , Australia , Niño , Diagnóstico Tardío , Diagnóstico Precoz , Luxación Congénita de la Cadera/diagnóstico por imagen , Luxación Congénita de la Cadera/terapia , Humanos , Lactante , Aparatos Ortopédicos , Evaluación de Resultado en la Atención de Salud , Resultado del Tratamiento
4.
J Pediatr Orthop ; 41(1): e50-e54, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32868515

RESUMEN

BACKGROUND: Bilateral slipped capital femoral epiphysis (SCFE) is common. The management of the contralateral hip in unilateral SCFE remains controversial. The aim of this study was to report on the clinical outcomes using a posterior sloping angle (PSA) threshold of 14.5 degrees for prophylactic fixation in preventing contralateral SCFE. METHODS: Having previously established through a retrospective study that PSA was predictive of future slip, the authors put in place a protocol where patients with unilateral SCFE who had a PSA ≥14.5 degrees on the contralateral side were offered prophylactic fixation. Those with unilateral SCFE presenting between January 2008 and December 2018 with a minimum of 12-month follow-up were included. Patients with renal or endocrine disorders were excluded. Primary outcomes were the number of slips prevented, the number needed to treat, and the complication rate. RESULTS: Of the 219 patients who were included, 114 (52.1%) underwent prophylactic fixation.A PSA threshold of 14.5 degrees prevented 77% of subsequent slips with a number needed to treat of 2.4 in our population. There were no cases of chondrolysis, avascular necrosis, or periprosthetic fracture associated with prophylactic pinning. CONCLUSIONS: Prophylactic fixation using a PSA of 14.5 degrees is safe, decreases unnecessary intervention, and reduces 77% of subsequent SCFE. The PSA can increase over time and the authors recommend that the protocol be applied for the duration of follow-up. LEVEL OF EVIDENCE: Level III.


Asunto(s)
Articulación de la Cadera/cirugía , Procedimientos Ortopédicos/estadística & datos numéricos , Epífisis Desprendida de Cabeza Femoral/prevención & control , Adolescente , Niño , Femenino , Humanos , Incidencia , Masculino , Nueva Gales del Sur/epidemiología , Estudios Prospectivos , Epífisis Desprendida de Cabeza Femoral/epidemiología , Epífisis Desprendida de Cabeza Femoral/cirugía
5.
J Orthop Sci ; 26(4): 684-689, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32713795

RESUMEN

BACKGROUND: Spine fusion is a common procedure for the treatment of severe scoliosis, a frequent and challenging deformity associated with Neurofibromatosis type 1 (NF1). Moreover, deficiencies in NF1-Ras-MEK signaling affect bone formation and resorption that in turn impacts on spine fusion outcomes. METHODS: In this study we describe a new model for AdCre virus induction of Nf1 deficiency in the spines of Nf1flox/flox mice. The virus is delivered locally to the mouse spine in a fusion procedure induced using BMP-2. Systemic adjunctive treatment with the MEK inhibitor (MEKi) PD0325901 and the bisphosphonate zoledronic acid (ZA) were next trialed in this model. RESULTS: AdCre delivery resulted in abundant fibrous tissue (Nf1null +393%, P < 0.001) and decreased marrow space (Nf1null -67%, P < 0.001) compared to controls. While this did not significantly impact on the bone volume of the fusion mass (Nf1null -14%, P = 0.999 n.s.), the presence of fibrous tissue was anticipated to impact on the quality of spine fusion. Multinucleated TRAP + cells were observed in the fibrous tissues seen in Nf1null spines. In Nf1null spines, MEKi increased bone volume (+194%, P < 0.001) whereas ZA increased bone density (+10%, P < 0.002) versus BMP-2 alone. Both MEKi and ZA decreased TRAP + cells in the fibrous tissue (MEKi -62%, P < 0.01; ZA -43%, P = 0.054). No adverse effects were seen with either MEKi or ZA treatment including weight loss or signs of illness or distress that led to premature euthanasia. CONCLUSIONS: These data not only support the utility of an AdCre-virus induced knockout spine model, but also support further investigation of MEKi and ZA as adjunctive therapies for improving BMP-2 induced spine fusion in the context of NF1.


Asunto(s)
Neurofibromatosis 1 , Animales , Benzamidas , Difenilamina/análogos & derivados , Modelos Animales de Enfermedad , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos , Neurofibromatosis 1/tratamiento farmacológico , Ácido Zoledrónico/farmacología
6.
Hum Mol Genet ; 27(4): 577-588, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29228356

RESUMEN

Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder with complex symptomology. In addition to a predisposition to tumors, children with NF1 can present with reduced muscle mass, global muscle weakness, and impaired motor skills, which can have a significant impact on quality of life. Genetic mouse models have shown a lipid storage disease phenotype may underlie muscle weakness in NF1. Herein we confirm that biopsy specimens from six individuals with NF1 similarly manifest features of a lipid storage myopathy, with marked accumulation of intramyocellular lipid, fibrosis, and mononuclear cell infiltrates. Intramyocellular lipid was also correlated with reductions in neurofibromin protein expression by western analysis. An RNASeq profile of Nf1null muscle from a muscle-specific Nf1 knockout mouse (Nf1MyoD-/-) revealed alterations in genes associated with glucose regulation and cell signaling. Comparison by lipid mass spectrometry demonstrated that Nf1null muscle specimens were enriched for long chain fatty acid (LCFA) containing neutral lipids, such as cholesterol esters and triacylglycerides, suggesting fundamentally impaired LCFA metabolism. The subsequent generation of a limb-specific Nf1 knockout mouse (Nf1Prx1-/-) recapitulated all observed features of human NF1 myopathy, including lipid storage, fibrosis, and muscle weakness. Collectively, these insights led to the evaluation of a dietary intervention of reduced LCFAs, and enrichment of medium-chain fatty acids (MCFAs) with L-carnitine. Following 8-weeks of dietary treatment, Nf1Prx1-/- mice showed a 45% increase in maximal grip strength, and a 71% reduction in intramyocellular lipid staining compared with littermates fed standard chow. These data link NF1 deficiency to fundamental shifts in muscle metabolism, and provide strong proof of principal that a dietary intervention can ameliorate symptoms.


Asunto(s)
Enfermedades Musculares/dietoterapia , Neurofibromatosis 1/dietoterapia , Adolescente , Adulto , Animales , Carnitina/uso terapéutico , Niño , Preescolar , Ácidos Grasos/uso terapéutico , Femenino , Humanos , Metabolismo de los Lípidos/fisiología , Masculino , Espectrometría de Masas , Ratones , Ratones Noqueados , Ratones Transgénicos , Debilidad Muscular/patología , Debilidad Muscular/terapia , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Calidad de Vida , Adulto Joven
7.
Calcif Tissue Int ; 106(2): 172-179, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31578632

RESUMEN

Clinical concerns have been raised over prior exposure to bisphosphonates impairing fracture healing. To model this, groups of male Wistar rats were assigned to saline control or treatment groups receiving 0.15 mg/kg (low dose), 0.5 mg/kg (medium dose), and 5 mg/kg (high dose) Pamidronate (PAM) twice weekly for 4 weeks. At this point, closed fractures were made using an Einhorn apparatus, and bisphosphonate dosing was continued until the experimental endpoint. Specimens were analyzed at 2 and 6 weeks (N = 8 per group per time point). Twice weekly PAM dosing was found to have no effect on early soft callus remodeling at 2 weeks post fracture. At this time point, the highest dose PAM group gave significant increases in bone volume (+ 10%, p < 0.05), bone mineral content (+ 30%, p < 0.01), and bone mineral density (+ 10%, p < 0.01). This PAM dosing regimen showed more substantive effects on hard callus at 6 weeks post fracture, with PAM treatment groups showing + 46-79% increased bone volume. Dynamic bone labeling showed reduced calcein signal in the PAM-treated calluses (38-63%, p < 0.01) and reduced MAR (32-49%, p < 0.01), suggesting a compensatory reduction in bone anabolism. These data support the concept that bisphosphonates lead to profound decreases in bone turnover in fracture repair, however, this does not affect soft callus remodeling.


Asunto(s)
Callo Óseo/efectos de los fármacos , Fracturas del Fémur/patología , Fracturas Cerradas/patología , Osteogénesis/efectos de los fármacos , Pamidronato/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/farmacología , Remodelación Ósea/efectos de los fármacos , Callo Óseo/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Fracturas del Fémur/tratamiento farmacológico , Curación de Fractura/efectos de los fármacos , Fracturas Cerradas/tratamiento farmacológico , Masculino , Tamaño de los Órganos/efectos de los fármacos , Osteogénesis Imperfecta/patología , Pamidronato/administración & dosificación , Ratas , Ratas Wistar , Factores de Tiempo
8.
J Cell Mol Med ; 23(3): 1735-1745, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30597759

RESUMEN

Osteogenesis imperfecta (OI) is commonly caused by heterozygous type I collagen structural mutations that disturb triple helix folding and integrity. This mutant-containing misfolded collagen accumulates in the endoplasmic reticulum (ER) and induces a form of ER stress associated with negative effects on osteoblast differentiation and maturation. Therapeutic induction of autophagy to degrade the mutant collagens could therefore be useful in ameliorating the ER stress and deleterious downstream consequences. To test this, we treated a mouse model of mild to moderate OI (α2(I) G610C) with dietary rapamycin from 3 to 8 weeks of age and effects on bone mass and mechanical properties were determined. OI bone mass and mechanics were, as previously reported, compromised compared to WT. While rapamycin treatment improved the trabecular parameters of WT and OI bones, the biomechanical deficits of OI bones were not rescued. Importantly, we show that rapamycin treatment suppressed the longitudinal and transverse growth of OI, but not WT, long bones. Our work demonstrates that dietary rapamycin offers no clinical benefit in this OI model and furthermore, the impact of rapamycin on OI bone growth could exacerbate the clinical consequences during periods of active bone growth in patients with OI caused by collagen misfolding mutations.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Colágeno Tipo I/fisiología , Modelos Animales de Enfermedad , Inmunosupresores/farmacología , Osteoblastos/efectos de los fármacos , Osteogénesis Imperfecta/tratamiento farmacológico , Sirolimus/farmacología , Animales , Apoptosis , Cadena alfa 1 del Colágeno Tipo I , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Osteoblastos/citología , Osteogénesis , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología
9.
Blood ; 129(26): 3452-3464, 2017 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-28515094

RESUMEN

Multiple myeloma (MM) is a plasma cell cancer that develops in the skeleton causing profound bone destruction and fractures. The bone disease is mediated by increased osteoclastic bone resorption and suppressed bone formation. Bisphosphonates used for treatment inhibit bone resorption and prevent bone loss but fail to influence bone formation and do not replace lost bone, so patients continue to fracture. Stimulating bone formation to increase bone mass and fracture resistance is a priority; however, targeting tumor-derived modulators of bone formation has had limited success. Sclerostin is an osteocyte-specific Wnt antagonist that inhibits bone formation. We hypothesized that inhibiting sclerostin would prevent development of bone disease and increase resistance to fracture in MM. Sclerostin was expressed in osteocytes from bones from naive and myeloma-bearing mice. In contrast, sclerostin was not expressed by plasma cells from 630 patients with myeloma or 54 myeloma cell lines. Mice injected with 5TGM1-eGFP, 5T2MM, or MM1.S myeloma cells demonstrated significant bone loss, which was associated with a decrease in fracture resistance in the vertebrae. Treatment with anti-sclerostin antibody increased osteoblast numbers and bone formation rate but did not inhibit bone resorption or reduce tumor burden. Treatment with anti-sclerostin antibody prevented myeloma-induced bone loss, reduced osteolytic bone lesions, and increased fracture resistance. Treatment with anti-sclerostin antibody and zoledronic acid combined increased bone mass and fracture resistance when compared with treatment with zoledronic acid alone. This study defines a therapeutic strategy superior to the current standard of care that will reduce fractures for patients with MM.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Fracturas Óseas/prevención & control , Osteocitos/química , Osteogénesis/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Animales , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Proteínas Morfogenéticas Óseas/inmunología , Línea Celular Tumoral , Difosfonatos/uso terapéutico , Marcadores Genéticos/inmunología , Humanos , Imidazoles/uso terapéutico , Ratones , Mieloma Múltiple/complicaciones , Células Tumorales Cultivadas , Ácido Zoledrónico
10.
Calcif Tissue Int ; 104(4): 426-436, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30535573

RESUMEN

Bone marrow transplantation (BMT) of healthy donor cells has been postulated as a strategy for treating osteogenesis imperfecta (OI) and other bone fragility disorders. The effect of engraftment by tail vein injection and/or marrow ablation by 6 Gy whole body irradiation were tested in Col1a2+/G610C (OI) mice as a model of mild-moderate OI. Dual-emission X-ray absorptiometry, microCT, and 4-point bending were used to measure bone volume (BV), bone mineral density (BMD), and biomechanical strength. BV, BMD, and mechanical strength were reduced in OI mice compared to wild type (WT) controls. BMT with and without irradiation yielded no difference in BV and BMD outcomes for both OI and WT mice, at 3 weeks. Transplantation of OI cells into OI mice to test for paracrine effects of BMT also showed no difference with non-transplanted OI mice. In a parallel cell tracking study, donor marrow was taken from transgenic mice constitutively expressing tdTomato and transplanted into WT mice. Lineage tracking demonstrated that irradiation considerably enhanced engraftment of tdTomato+ cells. However, tdTomato+ cells predominantly expressed TRAP and not AP, indicating engrafted donor cells were chiefly from the hematopoietic lineages. These data show that whole marrow transplantation fails to rescue the bone phenotype of Col1a2+/G610C (OI) mice and that osteopoietic engraftment is not significantly enhanced by irradiation. These findings are highly relevant to modern approaches focused on the gene repair of patient cells ex vivo and their subsequent reintroduction into the osteopoietic compartment via the circulation.


Asunto(s)
Trasplante de Médula Ósea , Huesos/metabolismo , Osteogénesis Imperfecta/terapia , Osteogénesis/fisiología , Animales , Densidad Ósea/fisiología , Trasplante de Médula Ósea/métodos , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteogénesis Imperfecta/genética
11.
Mol Genet Metab ; 123(4): 518-525, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29477258

RESUMEN

Neurofibromatosis Type 1 (NF1) is a common autosomal dominant genetic disorder While NF1 is primarily associated with predisposition for tumor formation, muscle weakness has emerged as having a significant impact on quality of life. NF1 inactivation is linked with a canonical upregulation Ras-MEK-ERK signaling. This in this study we tested the capacity of the small molecule MEK inhibitor PD0325901 to influence the intramyocellular lipid accumulation associated with NF1 deficiency. Established murine models of tissue specific Nf1 deletion in skeletal muscle (Nf1MyoD-/-) and limb mesenchyme (Nf1Prx1-/-) were tested. Developmental PD0325901 dosing of dams pregnant with Nf1MyoD-/- progeny rescued the phenotype of day 3 pups including body weight and lipid accumulation by Oil Red O staining. In contrast, PD0325901 treatment of 4 week old Nf1Prx1-/- mice for 8 weeks had no impact on body weight, muscle wet weight, activity, or intramyocellular lipid. Examination of day 3 Nf1Prx1-/- pups showed differences between the two tissue-specific knockout strains, with lipid staining greatest in Nf1MyoD-/- mice, and fibrosis higher in Nf1Prx1-/- mice. These data show that a MEK/ERK dependent mechanism underlies NF1 muscle metabolism during development. However, crosstalk from Nf1-deficient non-muscle mesenchymal cells may impact upon muscle metabolism and fibrosis in neonatal and mature myofibers.


Asunto(s)
Benzamidas/farmacología , Difenilamina/análogos & derivados , Extremidades/patología , Músculo Esquelético/patología , Enfermedades Musculares/prevención & control , Neurofibromatosis 1/fisiopatología , Neurofibromina 1/fisiología , Animales , Animales Recién Nacidos , Difenilamina/farmacología , Femenino , Proteínas de Homeodominio/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Proteína MioD/fisiología , Transducción de Señal , Proteínas ras/antagonistas & inhibidores , Proteínas ras/metabolismo
12.
Calcif Tissue Int ; 103(3): 298-310, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29845410

RESUMEN

Dickkopf-1 (DKK1) and sclerostin are antagonists of the Wnt/ß-catenin pathway and decreased expression of either results in increased bone formation and mass. As both affect the same signaling pathway, we aimed to elucidate the redundancy and/or compensation of sclerostin and DKK1. Weekly sclerostin antibody (Scl-Ab) was used to treat 9-week-old female Dkk1 KO (Dkk1-/-:Wnt3+/-) mice and compared to Scl-Ab-treated wild-type mice as well as vehicle-treated Dkk1 KO and wild-type animals. While Wnt3 heterozygote (Wnt3+/-) mice show no bone phenotype, Scl-Ab and vehicle-treated control groups of this genotype were included. Specimens were harvested after 3 weeks for microCT, bone histomorphometry, anti-sclerostin immunohistochemistry, and biomechanical testing. Scl-Ab enhanced bone anabolism in all treatment groups, but with synergistic enhancement seen in the cancellous compartment of Dkk1 KO mice (bone volume + 55% Dkk1 KO p < 0.01; + 22% wild type p < 0.05). Scl-Ab treatment produced less marked increases in cortical bone of the tibiae, with anabolic effects similar across genotypes. Mechanical testing confirmed that Scl-Ab improved strength across all genotypes; however, no enhancement was seen within Dkk1 KO mice. Dynamic bone labeling showed that Scl-Ab treatment was associated with increased bone formation, regardless of genotype. Immunohistochemical staining for sclerostin protein indicated no differences in the Dkk1 KO mice, indicating that the increased Wnt signaling associated with DKK1 deficiency was not compensated by upregulation of sclerostin protein. These data suggest complex interactions between Wnt signaling factors in bone, but critically illustrate synergy between DKK1 deficiency and Scl-Ab treatment. These data support the application of dual-targeted therapeutics in the modulation of bone anabolism.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/fisiología , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Osteogénesis/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Anticuerpos Monoclonales/farmacología , Femenino , Ratones , Ratones Noqueados , Osteogénesis/efectos de los fármacos
13.
Calcif Tissue Int ; 102(1): 105-116, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29105022

RESUMEN

Wnt antagonist Dkk1 is a negative regulator of bone formation and Dkk1 +/- heterozygous mice display a high bone mass phenotype. Complete loss of Dkk1 function disrupts embryonic head development. Homozygous Dkk1 -/- mice that were heterozygous for Wnt3 loss of function mutation (termed Dkk1 KO) are viable and allowed studying the effects of homozygous inactivation of Dkk1 on bone formation. Dkk1 KO mice showed a high bone mass phenotype exceeding that of heterozygous mice as well as a high incidence of polydactyly and kinky tails. Whole body bone density was increased in the Dkk1 KO mice as shown by longitudinal dual-energy X-ray absorptiometry. MicroCT analysis of the distal femur revealed up to 3-fold increases in trabecular bone volume and up to 2-fold increases in the vertebrae, compared to wild type controls. Cortical bone was increased in both the tibiae and vertebrae, which correlated with increased strength in tibial 4-point bending and vertebral compression tests. Dynamic histomorphometry identified increased bone formation as the mechanism underlying the high bone mass phenotype in Dkk1 KO mice, with no changes in bone resorption. Mice featuring only Wnt3 heterozygosity showed no evident bone phenotype. Our findings highlight a critical role for Dkk1 in the regulation of bone formation and a gene dose-dependent response to loss of DKK1 function. Targeting Dkk1 to enhance bone formation offers therapeutic potential for osteoporosis.


Asunto(s)
Densidad Ósea/fisiología , Huesos/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Osteogénesis/genética , Animales , Enfermedades Óseas Metabólicas/metabolismo , Resorción Ósea/metabolismo , Huesos/patología , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones Noqueados , Fenotipo
14.
Clin Orthop Relat Res ; 476(6): 1311-1323, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29698291

RESUMEN

BACKGROUND: Infection of open fractures remains a significant cause of morbidity and mortality to patients worldwide. Early administration of prophylactic antibiotics is known to improve outcomes; however, increasing concern regarding antimicrobial resistance makes finding new compounds for use in such cases a pressing area for further research. CSA-90, a synthetic peptidomimetic compound, has previously demonstrated promising antimicrobial action against Staphylococcus aureus in rat open fractures. However, its efficacy against antibiotic-resistant microorganisms, its potential as a therapeutic agent in addition to its prophylactic effects, and its proosteogenic properties all require further investigation. QUESTIONS/PURPOSES: (1) Does prophylactic treatment with CSA-90 reduce infection rates in a rat open fracture model inoculated with S aureus, methicillin-resistant S aureus (MRSA), and methicillin-resistant Staphylococcus epidermidis (MRSE) as measured by survival, radiographic union, and deep tissue swab cultures? (2) Does CSA-90 reduce infection rates when administered later in the management of an open fracture as measured by survival, radiographic union, and deep tissue swab cultures? (3) Does CSA-90 demonstrate a synergistic proosteogenic effect with bone morphogenetic protein 2 (BMP-2) in a noninfected rat ectopic bone formation assay as assessed by micro-CT bone volume measurement? (4) Can CSA-90 elute and retain its antimicrobial efficacy in vitro when delivered using clinically relevant agents measured using a Kirby-Bauer disc diffusion assay? METHODS: All in vivo studies were approved by the local animal ethics committee. In the open fracture studies, 12-week-old male Wistar rats underwent open midshaft femoral fractures stabilized with a 1.1-mm Kirschner wire and 10 µg BMP-2 ± 500 µg CSA-90 was applied to the fracture site using a collagen sponge along with 1 x 10 colony-forming units of bacteria (S aureus/MRSA/MRSE; n = 10 per group). In the delayed treatment study, débridement and treatment with 500 µg CSA-90 were performed at Day 1 and Day 5 after injury and bacterial insult (S aureus). All animals were reviewed daily for signs of local infection and/or sepsis. An independent, blinded veterinarian reviewed twice-weekly radiographs, and rats showing osteolysis and/or declining overall health were culled at his instruction. The primary outcome of both fracture studies was fracture infection, incorporating survival, radiographic union, and deep tissue swab cultures. For the ectopic bone formation assay, 0 to 10 µg BMP-2 and 0 to 500 µg CSA-90 were delivered on a collagen sponge into bilateral quadriceps muscle pouches of 8-week-old rats (n = 10 per group). Micro-CT quantification of bone volume and descriptive histologic analysis were performed for all in vivo studies. Modified Kirby-Bauer disc diffusion assays were used to quantify antimicrobial activity in vitro using four different delivery methods, including bone cement. RESULTS: Infection was observed in none of the MRSA inoculated open fractures treated with CSA-90 with 10 of 10 deep tissue swab cultures negative at the time of cull. Median survival was 43 days (range, 11-43 days) in the treated group versus 11 days (range, 8-11 days) in the untreated MRSA inoculated group (p < 0.001). However, delayed débridement and treatment of open fractures with CSA-90 at either Day 1 or Day 5 did not prevent infection, resulting in early culls by Day 21 with positive swab cultures (10 of 10 for each time point). Maximal ectopic bone formation was achieved with 500 µg CSA-90 and 10 µg BMP-2 (mean volume, 9.58 mm; SD, 7.83), creating larger bone nodules than formed with 250 µg CSA-90 and 10 µg BMP-2 (mean volume, 1.7 mm; SD, 1.07; p < 0.001). Disc diffusion assays showed that CSA-90 could successfully elute from four potential delivery agents including calcium sulphate (mean zone of inhibition, 11.35 mm; SD, 0.957) and bone cement (mean, 4.67 mm; SD, 0.516). CONCLUSIONS: CSA-90 shows antimicrobial action against antibiotic-resistant Staphylococcal strains in vitro and in an in vivo model of open fracture infection. CLINICAL RELEVANCE: The antimicrobial properties of CSA-90 combined with further evidence of its proosteogenic potential make it a promising compound to develop further for orthopaedic applications.


Asunto(s)
Profilaxis Antibiótica/métodos , Fracturas del Fémur/tratamiento farmacológico , Fracturas Abiertas/tratamiento farmacológico , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Pregnanos/farmacología , Propilaminas/farmacología , Infecciones Estafilocócicas/prevención & control , Animales , Modelos Animales de Enfermedad , Fracturas del Fémur/microbiología , Fracturas Abiertas/microbiología , Masculino , Ratas , Ratas Wistar , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/efectos de los fármacos
15.
Calcif Tissue Int ; 101(2): 217-228, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28391431

RESUMEN

Open fractures remain a challenge in orthopedics. Current strategies to intervene are often inadequate, particularly in severe fractures or when treatment is delayed. Sclerostin is a negative regulator of bone growth and sclerostin-neutralizing antibodies (Scl-Ab) can increase bone mass and strength. The application of these antibodies to improve orthopedic repair has shown varied results, and may be dependent on the location and severity of the bony injury. We examined Scl-Ab treatment within an established rat osteotomy model with periosteal stripping analogous to open fracture repair. In one study, Scl-Ab was given 25 mg/kg bi-weekly, either from the time of fracture or from 3 weeks post-fracture up to an end-point of 12 weeks. A second study treated only delayed union open fractures that did not show radiographic union by week 6 post-fracture. Outcome measures included radiographic union, microCT analysis of bone volume and architecture, and histology. In the first study, Scl-Ab given from either 0 or 3 weeks significantly improved callus bone volume (+52%, p < 0.05 and +58%, p < 0.01) at 12 weeks, as well as strength (+48%, p < 0.05 and +70%, p < 0.05). Despite these improvements, union rate was not changed. In the second study treating only established delayed fractures, bony callus volume was similarly increased by Scl-Ab treatment; however, this did not translate to increased biomechanical strength or union improvement. Sclerostin antibody treatment has limited effects on the healing of challenging open fractures with periosteal stripping, but shows the greatest benefits on callus size and strength with earlier intervention.


Asunto(s)
Anticuerpos/farmacología , Densidad Ósea/efectos de los fármacos , Proteínas Morfogenéticas Óseas/inmunología , Callo Óseo/patología , Marcadores Genéticos/inmunología , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Modelos Animales de Enfermedad , Fracturas del Fémur/tratamiento farmacológico , Fracturas del Fémur/patología , Fémur/efectos de los fármacos , Fémur/patología , Curación de Fractura/efectos de los fármacos , Masculino , Osteogénesis/efectos de los fármacos , Osteotomía/métodos , Ratas
16.
Biomacromolecules ; 18(6): 1736-1746, 2017 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-28535038

RESUMEN

Biodegradable polymers are appealing material for the manufacturing of surgical implants as such implants break down in vivo, negating the need for a subsequent operation for removal. Many biocompatible polymers produce acidic breakdown products that can lead to localized inflammation and osteolysis. This study assesses the feasibility of fabricating implants out of poly(propylene carbonate) (PPC)-starch that degrades into CO2 and water. The basic compression modulus of PPC-starch (1:1 w/w) is 34 MPa; however, the addition of glycerol (1% w/w) and water as plasticizers doubles this value and enhances the surface wettability. The bioactivity and stiffness of PPC-starch blends is increased by the addition of bioglass microparticles (10% w/w) as shown by in vitro osteoblast differentiation assay and mechanical testing. MicroCT analysis confirms that the bioglass microparticles are evenly distributed throughout biomaterial. PPC-starch-bioglass was tested in vivo in two animal models. A murine subcutaneous pellet degradation assay demonstrates that the PPC-starch-bioglass blend's volume fraction loss is 46% after 6 months postsurgery, while it is 27% for poly(lactic acid). In a rat knee implantation model, PPC-starch-bioglass screws inserted into the distal femur show osseointegration with no localized adverse effects after 3 and 12 weeks. These data support the further development of PPC-starch-bioglass as a medical biomaterial.


Asunto(s)
Implantes Absorbibles , Materiales Biocompatibles/síntesis química , Interfase Hueso-Implante/fisiología , Cerámica/farmacología , Polipropilenos/síntesis química , Almidón/química , Animales , Materiales Biocompatibles/metabolismo , Materiales Biocompatibles/farmacología , Interfase Hueso-Implante/anatomía & histología , Interfase Hueso-Implante/diagnóstico por imagen , Dióxido de Carbono/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Cerámica/química , Femenino , Fémur/cirugía , Glicerol/química , Glicerol/metabolismo , Humanos , Hidrólisis , Masculino , Ratones , Ratones Endogámicos BALB C , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Polipropilenos/metabolismo , Polipropilenos/farmacología , Ratas , Almidón/metabolismo , Agua/metabolismo , Humectabilidad
17.
Hum Mol Genet ; 23(5): 1250-9, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24163128

RESUMEN

There is emerging evidence for reduced muscle function in children with neurofibromatosis type 1 (NF1). We have examined three murine models featuring NF1 deficiency in muscle to study the effect on muscle function as well as any underlying pathophysiology. The Nf1(+/-) mouse exhibited no differences in overall weight, lean tissue mass, fiber size, muscle weakness as measured by grip strength or muscle atrophy-recovery with limb disuse, although this model lacks many other characteristic features of the human disease. Next, muscle-specific knockout mice (Nf1muscle(-/-)) were generated and they exhibited a failure to thrive leading to neonatal lethality. Intramyocellular lipid accumulations were observed by electron microscopy and Oil Red O staining. More mature muscle specimens lacking Nf1 expression taken from the limb-specific Nf1Prx1(-/-) conditional knockout line showed a 10-fold increase in muscle triglyceride content. Enzyme assays revealed a significant increase in the activities of oxidative metabolism enzymes in the Nf1Prx1(-/-) mice. Western analyses showed increases in the expression of fatty acid synthase and the hormone leptin, as well as decreased expression of a number of fatty acid transporters in this mouse line. These data support the hypothesis that NF1 is essential for normal muscle function and survival and are the first to suggest a direct link between NF1 and mitochondrial fatty acid metabolism.


Asunto(s)
Desarrollo de Músculos/genética , Músculos/metabolismo , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Animales , Peso Corporal , Resorción Ósea/genética , Huesos/metabolismo , Huesos/patología , Modelos Animales de Enfermedad , Genes Letales , Heterocigoto , Homocigoto , Humanos , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Fuerza Muscular/genética , Músculos/patología , Músculos/ultraestructura , Neurofibromina 1/deficiencia , Tamaño de los Órganos
18.
Paediatr Anaesth ; 26(12): 1188-1196, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27663858

RESUMEN

BACKGROUND: Vascular complications following pediatric liver transplantation occur in 8-10% of cases, and no continuous, non-invasive monitoring for this problem exists. Near infrared spectroscopy (NIRS) allows non-invasive, continuous, transcutaneous assessment of hemoglobin oxygenation (StO2 ) 1-4 cm below the skin surface. AIMS: We hypothesized that transcutaneous NIRS would be able to detect severe hepatic ischemia, and tested this in an animal model using 15-20 kg and 5-7 kg juvenile pigs. MATERIALS AND METHODS: Direct liver surface and transcutaneous hepatic tissue hemoglobin oxygen saturation (StO2 ) were measured during occlusions of the hepatic artery and portal vein. Changes in hepatic delivery of oxygen (HepDO2 ) were calculated for each ischemic challenge and compared to changes in direct liver surface (DirHepStO2 ) and transcutaneous liver StO2 measurements (CutHepStO2 ). RESULTS: In the 15-20 kg animals during complete occlusion, CutHepStO2 decreased by 6.0(±4.9)%, whilst DirHepStO2 decreased by 83.7(±7.2)%. In the 5-7 kg animals during complete occlusion, CutHepStO2 decreased by 27.4(±8.5)%, whilst DirHepStO2 decreased by 82.8(±4.6)%. CONCLUSION: Transcutaneous hepatic StO2 monitoring cannot reliably detect severe hepatic ischemia in a juvenile porcine model, although a stronger and potentially useful signal is seen in 5-7 kg pigs. Trials of this technology should be currently restricted to situations where the organ is less than 1 cm from the skin surface, corresponding to infants of <10 kg.


Asunto(s)
Isquemia/diagnóstico por imagen , Hígado/irrigación sanguínea , Hígado/diagnóstico por imagen , Espectroscopía Infrarroja Corta/métodos , Animales , Modelos Animales de Enfermedad , Masculino , Reproducibilidad de los Resultados , Porcinos
19.
J Pediatr Orthop ; 36(6): 651-5, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25955168

RESUMEN

BACKGROUND: The posterior sloping angle (PSA) has been shown to be an objective and reproducible predictor of the risk of patients developing contralateral slipped capital femoral epiphysis (SCFE); however, prophylactic fixation remains controversial. This in vitro study investigates the biomechanical basis of using a 15-degree PSA as a threshold for prophylactic fixation. METHODS: Synthetic bone in vitro models of the proximal femur were constructed with a PSA of 10 degrees as a control (normal) group (n=6) by performing an osteotomy at the physis and gluing the head back onto the neck. SCFE groups were created with a PSA of 15, 20, 25, 30, 50, or 60 degrees, by excising a wedge from the posterior neck and gluing them back at the new angle with corresponding posterior translation proportional to the slip angle, and loaded superoinferiorly in compression, to failure. Ultimate strength, energy to failure, and stiffness were recorded. RESULTS: Increasing the PSA from 10 to 15 degrees only reduced ultimate strength by 13% (P>0.05; CI, -0.21 to -0.06), though a significantly lesser energy to failure was required (-58%, P<0.05; CI, -0.68 to -0.48). Increasing the angle to 20 degrees resulted in a further significant decrease in strength (-19%, P<0.05; CI, -0.28 to -0.10) and energy to failure (-45%, P<0.05; CI, -0.53 to -0.84). The severe SCFE (60-degree PSA) was significantly weaker and less rigid that the control, and the mild and moderate SCFE models (P<0.01). CONCLUSIONS: These biomechanical data support the threshold of 15-degree PSA as an objective measure for prophylactic fixation of the contralateral hip in SCFE. CLINICAL RELEVANCE: The number needed to treat with (minimally invasive) prophylactic fixation to prevent contralateral SCFE can be minimized if the above-mentioned threshold is used.


Asunto(s)
Artrometría Articular/métodos , Fijación de Fractura/métodos , Epífisis Desprendida de Cabeza Femoral , Fenómenos Biomecánicos , Fémur/diagnóstico por imagen , Placa de Crecimiento , Articulación de la Cadera/diagnóstico por imagen , Humanos , Modelos Anatómicos , Radiografía/métodos , Epífisis Desprendida de Cabeza Femoral/diagnóstico , Epífisis Desprendida de Cabeza Femoral/fisiopatología , Epífisis Desprendida de Cabeza Femoral/cirugía
20.
Dev Growth Differ ; 57(1): 10-23, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25389084

RESUMEN

Mouse models incorporating inducible Cre-ERT2/LoxP recombination coupled with sensitive fluorescent reporter lines are being increasingly used to track cell lineages in vivo. In this study we use two inducible reporter strains, Ai9iCol2a1 (Ai9×Col2a1-creERT2) to track contribution of chondrogenic progenitors during bone regeneration in a closed fracture model and Ai9i UBC (Ai9×UBC-creERT2) to examine methods for inducing localized recombination. By comparing with Ai9 littermate controls as well as inducible reporter mice not dosed with tamoxifen, we revealed significant leakiness of the CreERT2 system, particularly in the bone marrow of both lines. These studies highlight the challenges associated with highly sensitive reporters that may be activated without induction in tissues where the CreERT2 fusion is expressed. Examination of the growth plate in the Ai9iCol2a1 strain showed cells of the osteochondral lineage (cell co-staining with chondrocyte and osteoblast markers) labeled with the tdTom reporter. However, no such labeling was noted in healing fractures of Ai9iCol2a1 mice. Attempts to label a single limb using intramuscular injection of 4-hydroxytamoxifen in the Ai9i UBC strain resulted in complete labeling of the entire animal, comparable to intraperitoneal injection. While a challenge to interpret, these data are nonetheless informative regarding the limitations of these inducible reporter models, and justify caution and expansive controls in future studies using such models.


Asunto(s)
Rastreo Celular/métodos , Condrocitos/metabolismo , Curación de Fractura/fisiología , Fracturas Óseas/metabolismo , Genes Reporteros , Osteoblastos/metabolismo , Animales , Condrocitos/patología , Femenino , Fracturas Óseas/patología , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA