Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.536
Filtrar
1.
PLoS One ; 19(5): e0303752, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753866

RESUMEN

BACKGROUND: First metatarsophalangeal joint arthrodesis is a typical medical treatment performed in cases of arthritis or joint deformity. The gold standard for this procedure is arthrodesis stabilisation with the dorsally positioned plate. However, according to the authors' previous studies, medially positioned plate provides greater bending stiffness. It is worth to compare the mechanical conditions for bone formation in the fracture callus for both placements of the locking plate. METHODS: Two finite element models of the first metatarsophalangeal joint with the dorsally and medially positioned plate were defined in the Abaqus software to simulate differentiation of the fracture callus. A simplified load application, i.e. one single step per each day and the diffusion of the mesenchymal stem cells into the fracture region were assumed in an iterative hardening process. The changes of the mesenchymal stem cells into different phenotypes during the callus stiffening were governed by the octahedral shear strain and interstitial fluid velocity according to Prendergast mechanoregulation theory. Basing on the obtained results the progress of the cartilage and bone tissues formation and their distribution within the callus were compared between two models. FINDINGS: The obtained results suggest that after 6 weeks of simulation the healing progress is in general comparable for both plates. However, earlier closing of external callus was observed for the medially positioned plate which had greater vertical bending stiffness. This process enables faster internal callus hardening and promotes symmetrical bridging.


Asunto(s)
Artrodesis , Placas Óseas , Análisis de Elementos Finitos , Articulación Metatarsofalángica , Artrodesis/métodos , Humanos , Articulación Metatarsofalángica/cirugía , Fenómenos Biomecánicos , Callo Óseo , Curación de Fractura
2.
J Am Soc Mass Spectrom ; 35(6): 1184-1196, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38679918

RESUMEN

Bone fracture healing is a complex process in which specific molecular knowledge is still lacking. The citrulline-arginine-nitric oxide metabolism is one of the involved pathways, and its enrichment via citrulline supplementation can enhance fracture healing. This study investigated the molecular effects of citrulline supplementation during the different fracture healing phases in a rat model. Microcomputed tomography (µCT) was applied for the analysis of the fracture callus formation. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) and liquid-chromatography tandem mass spectrometry (LC-MS/MS) were used for lipid and protein analyses, respectively. µCT analysis showed no significant differences in the fracture callus volume and volume fraction between the citrulline supplementation and control group. The observed lipid profiles for the citrulline supplementation and control group were distinct for the different fracture healing stages. The main contributing lipid classes were phosphatidylcholines (PCs) and lysophosphatidylcholines (LPCs). The changing effect of citrulline supplementation throughout fracture healing was indicated by changes in the differentially expressed proteins between the groups. Pathway analysis showed an enhancement of fracture healing in the citrulline supplementation group in comparison to the control group via improved angiogenesis and earlier formation of the soft and hard callus. This study showed the molecular effects on lipids, proteins, and pathways associated with citrulline supplementation during bone fracture healing, even though no effect was visible with µCT.


Asunto(s)
Citrulina , Curación de Fractura , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Microtomografía por Rayos X , Animales , Curación de Fractura/efectos de los fármacos , Ratas , Citrulina/análisis , Citrulina/metabolismo , Citrulina/farmacología , Espectrometría de Masas en Tándem/métodos , Microtomografía por Rayos X/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Suplementos Dietéticos/análisis , Modelos Animales de Enfermedad , Masculino , Callo Óseo/efectos de los fármacos , Callo Óseo/diagnóstico por imagen , Callo Óseo/metabolismo , Cromatografía Liquida/métodos , Lisofosfatidilcolinas/metabolismo , Lisofosfatidilcolinas/análisis , Fosfatidilcolinas/metabolismo , Fosfatidilcolinas/análisis , Fosfatidilcolinas/farmacología
3.
Bone ; 183: 117091, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38570121

RESUMEN

The healing of bone fractures can become aberrant and lead to nonunions which in turn have a negative impact on patient health. Understanding why a bone fails to normally heal will enable us to make a positive impact in a patient's life. While we have a wealth of molecular data on rodent models of fracture repair, it is not the same with humans. As such, there is still a lack of information regarding the molecular differences between normal physiological repair and nonunions. This study was designed to address this gap in our molecular knowledge of the human repair process by comparing differentially expressed genes (DEGs) between physiological fracture callus and two different nonunion types, hypertrophic (HNU) and oligotrophic (ONU). RNA sequencing data revealed over ∼18,000 genes in each sample. Using the physiological callus as the control and the nonunion samples as the experimental groups, bioinformatic analyses identified 67 and 81 statistically significant DEGs for HNU and ONU, respectively. Out of the 67 DEGs for the HNU, 34 and 33 were up and down-regulated, respectively. Similarly, out of the 81 DEGs for the ONU, 48 and 33 were up and down-regulated, respectively. Additionally, we also identified common genes between the two nonunion samples; 8 (10.8 %) upregulated and 12 (22.2 %) downregulated. We further identified many biological processes, with several statistically significant ones. Some of these were related to muscle and were common between the two nonunion samples. This study represents the first comprehensive attempt to understand the global molecular events occurring in human nonunion biology. With further research, we can perhaps decipher new molecular pathways involved in aberrant healing of human bone fractures that can be therapeutically targeted.


Asunto(s)
Fracturas Óseas , Fracturas no Consolidadas , Humanos , Curación de Fractura/fisiología , Fracturas no Consolidadas/metabolismo , Callo Óseo/metabolismo , Expresión Génica
4.
Eur J Orthop Surg Traumatol ; 34(4): 2147-2153, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38564013

RESUMEN

INTRODUCTION: Distal femur fractures account for 3-6% of all femur fractures. Internal fixation of most distal femur fractures with an anatomic lateral locking plate should permit some motion at the metaphyseal portion of the fracture when secondary bone healing is planned by the operating surgeon. While several studies have been performed evaluating union rates for distal femur fractures with stainless steel and titanium plates, the timing of callus formation between stainless steel and titanium implants used as bridge plates for distal femur fractures (AO/OTA 33-A and -C) has been investigated to a lesser extent. We hypothesize that callus will be visualized earlier with post-operative radiographs with titanium versus stainless steel bridge plates. METHODS: We retrospectively reviewed a consecutive cohort of patients over 18 years of age with acute AO/OTA 33-A and 33-C fracture patterns treated with an isolated stainless steel or titanium lateral bridge plate within 4 weeks of injury by a single fellowship-trained orthopedic trauma surgeon from 2011 to 2020 at one academic Level 1 trauma center. An independent, fellowship-trained orthopedic trauma attending surgeon reviewed anterior-posterior (AP) and lateral radiographs from every available post-operative clinic visit and graded them using the Modified Radiographic Score for Tibia (mRUST). RESULTS: Twenty-five subjects were included in the study with 10 with stainless steel and 15 with titanium plates. There were no significant differences in demographics between both groups, including age, sex, BMI, injury classification, open versus closed, mechanism, and laterality. Statistically significant increased mRUST scores, indicating increased callus formation, were seen on 12-week radiographs (8.4 vs. 11.9, p = 0.02) when titanium bridge plates were used. There were no statistically significant differences in mRUST scores at 6 or 24-weeks, but scores in the titanium group were higher in at every timepoint. DISCUSSION: In conclusion, we observed greater callus formation at 12 weeks after internal fixation of 33-A and 33-C distal femur fractures treated with titanium locked lateral distal femoral bridge plates compared to stainless steel plates. Our data suggest that titanium metallurgy may have quicker callus formation compared to stainless steel if an isolated, lateral locked bridge plate is chosen for distal femur fracture fixation.


Asunto(s)
Placas Óseas , Callo Óseo , Fracturas del Fémur , Fijación Interna de Fracturas , Acero Inoxidable , Titanio , Humanos , Fracturas del Fémur/cirugía , Fracturas del Fémur/diagnóstico por imagen , Fracturas del Fémur/fisiopatología , Estudios Retrospectivos , Fijación Interna de Fracturas/instrumentación , Fijación Interna de Fracturas/métodos , Masculino , Callo Óseo/diagnóstico por imagen , Femenino , Persona de Mediana Edad , Adulto , Radiografía , Curación de Fractura/fisiología , Anciano , Fracturas Femorales Distales
5.
Cells ; 13(6)2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38534368

RESUMEN

Fracture callus formation is a dynamic stage of bone activity and repair with precise, spatially localized gene expression. Metastatic breast cancer impairs fracture healing by disrupting bone homeostasis and imparting an altered genomic profile. Previous sequencing techniques such as single-cell RNA and in situ hybridization are limited by missing spatial context and low throughput, respectively. We present a preliminary approach using the Visium CytAssist spatial transcriptomics platform to provide the first spatially intact characterization of genetic expression changes within an orthopedic model of impaired fracture healing. Tissue slides prepared from BALB/c mice with or without MDA-MB-231 metastatic breast cancer cells were used. Both unsupervised clustering and histology-based annotations were performed to identify the hard callus, soft callus, and interzone for differential gene expression between the wild-type and pathological fracture model. The spatial transcriptomics platform successfully localized validated genes of the hard (Dmp1, Sost) and soft callus (Acan, Col2a1). The fibrous interzone was identified as a region of extensive genomic heterogeneity. MDA-MB-231 samples demonstrated downregulation of the critical bone matrix and structural regulators that may explain the weakened bone structure of pathological fractures. Spatial transcriptomics may represent a valuable tool in orthopedic research by providing temporal and spatial context.


Asunto(s)
Callo Óseo , Fracturas del Fémur , Ratones , Animales , Callo Óseo/metabolismo , Callo Óseo/patología , Fracturas del Fémur/patología , Curación de Fractura , Perfilación de la Expresión Génica
6.
Front Immunol ; 15: 1352819, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38455063

RESUMEN

Bone fracture repair is a complex, multi-step process that involves communication between immune and stromal cells to coordinate the repair and regeneration of damaged tissue. In the US, 10% of all bone fractures do not heal properly without intervention, resulting in non-union. Complications from non-union fractures are physically and financially debilitating. We now appreciate the important role that immune cells play in tissue repair, and the necessity of the inflammatory response in initiating healing after skeletal trauma. The temporal dynamics of immune and stromal cell populations have been well characterized across the stages of fracture healing. Recent studies have begun to untangle the intricate mechanisms driving the immune response during normal or atypical, delayed healing. Various in vivo models of fracture healing, including genetic knockouts, as well as in vitro models of the fracture callus, have been implemented to enable experimental manipulation of the heterogeneous cellular environment. The goals of this review are to (1): summarize our current understanding of immune cell involvement in fracture healing (2); describe state-of-the art approaches to study inflammatory cells in fracture healing, including computational and in vitro models; and (3) identify gaps in our knowledge concerning immune-stromal crosstalk during bone healing.


Asunto(s)
Curación de Fractura , Fracturas Óseas , Humanos , Callo Óseo , Células del Estroma , Comunicación Celular
7.
Med Eng Phys ; 125: 104122, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38508793

RESUMEN

The embracing fixator is one of the widely used internal fixation implants for bone fracture treatment. However, the stress shielding effect, a stress imbalance between the implant and bone caused by the mismatch in mechanical properties between them, is a significant and critical issue that may lead to treatment failure. Thus, it is of great importance to design the implant with an appropriate stiffness which can mitigate the stress shielding effect and provide the most favorable mechanical environment for bone healing and remodeling. To this end, a time-dependent topology optimization algorithm considering bone remodeling is proposed to optimize an embracing fixator used in the tibia fracture treatment. The change of callus density over time is simulated based on a bone remodeling model, and the callus density after a period of bone remodeling is selected to be the design objective to maximize. The design constraints include volume and the compliance of the whole fixation system. Meanwhile, the influence of the constraints on the regularity of material distribution of the optimized result is also studied. Besides, to test the effectiveness of the consideration of the bone remodeling in the embracing fixator design, a topology optimization concerning the minimization of the compliance of the entire system is also performed to make a comparison. Finally, the safety performance of optimized results considering bone remodeling is also verified by static analysis.


Asunto(s)
Prótesis e Implantes , Fracturas de la Tibia , Humanos , Remodelación Ósea , Callo Óseo , Fijación Interna de Fracturas/métodos , Fracturas de la Tibia/cirugía , Curación de Fractura
8.
Turk Neurosurg ; 34(2): 314-324, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38497185

RESUMEN

AIM: To examine the effect of cerebellar damage on the process of fracture healing. MATERIAL AND METHODS: A total of forty-two male rats were selected at random and subsequently allocated into three distinct groups. The experimentals were divided into two subgroups within each group, with the intention of sacrificing them during the third and sixth weeks. Group 1 had isolated femoral fracture, Group 2 had femoral fracture after craniotomy, and Group 3 had femoral fracture accompanying cerebellar injury after craniotomy. Left femoral fractures in rats in all groups were treated using an intramedullary Kirschner wire. Radiological, histological, and biochemical evaluations were conducted at 3 and 6 weeks to assess the processes of fracture healing. To determine the effects of fracture healing and cerebellar injury on oxidant-antioxidant systems, catalase (CAT), malondialdehyde, superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities were measured. RESULTS: Between the time frame of 3 to 6 weeks, Group 3 had higher radiography scores, alkaline phosphatase levels, callus/ diaphyse ratio, callus improvement, and bone mineral density in comparison to the other groups. The activity of SOD was found to be statistically negligible in all groups, suggesting that SOD does not have a substantial impact on fracture healing in cerebellar injury. However, notable increases in the activity of GPx and CAT enzymes were observed, showing their considerable involvement in the process of fracture healing. CONCLUSION: Cerebellar injury reduces the oxidative stress in the fracture area and contributes positively to fracture healing by means of radiologically, biochemically and histopathologically.


Asunto(s)
Fracturas del Fémur , Curación de Fractura , Ratas , Animales , Masculino , Callo Óseo/metabolismo , Callo Óseo/patología , Fracturas del Fémur/metabolismo , Fracturas del Fémur/patología , Fracturas del Fémur/cirugía , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Antioxidantes/farmacología
9.
Dev Cell ; 59(9): 1192-1209.e6, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38554700

RESUMEN

Bone is regarded as one of few tissues that heals without fibrous scar. The outer layer of the periosteum is covered with fibrous tissue, whose function in bone formation is unknown. We herein developed a system to distinguish the fate of fibrous-layer periosteal cells (FL-PCs) from the skeletal stem/progenitor cells (SSPCs) in the cambium-layer periosteum and bone marrow in mice. We showed that FL-PCs did not participate in steady-state osteogenesis, but formed the main body of fibrocartilaginous callus during fracture healing. Moreover, FL-PCs invaded the cambium-layer periosteum and bone marrow after fracture, forming neo-SSPCs that continued to maintain the healed bones throughout adulthood. The FL-PC-derived neo-SSPCs expressed lower levels of osteogenic signature genes and displayed lower osteogenic differentiation activity than the preexisting SSPCs. Consistent with this, healed bones were thinner and formed more slowly than normal bones. Thus, the fibrous periosteum becomes the cellular origin of bones after fracture and alters bone properties permanently.


Asunto(s)
Diferenciación Celular , Curación de Fractura , Fracturas Óseas , Osteogénesis , Periostio , Animales , Periostio/metabolismo , Ratones , Osteogénesis/fisiología , Curación de Fractura/fisiología , Fracturas Óseas/patología , Fracturas Óseas/metabolismo , Células Madre/metabolismo , Células Madre/citología , Ratones Endogámicos C57BL , Callo Óseo/metabolismo , Callo Óseo/patología , Masculino
10.
Biochem Biophys Res Commun ; 703: 149634, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38354465

RESUMEN

Fractures are frequent and severe musculoskeletal injuries. This study aimed to investigate the function of tenascin-C (TNC) in regulating chondrogenic during fracture healing and elucidate the underlying molecular mechanisms. A well-established femur fracture model in male C57BL/6J mice was used to transect the middle diaphysis of the femur. To identify the essential role of TNC, shTNC lentiviruses or TNC protein were administered in the animal model. Micro-CT analysis, histologic analysis, immunostaining assays, and gene expression analysis were employed to investigate the effect of TNC during fracture healing. An in vitro mesenchymal stem cell culture system was developed to investigate the role and molecular mechanism of TNC in regulating chondrogenesis. TNC expression was induced at the inflammatory phase and peaked at the cartilaginous callus phase during fracture healing. Knockdown of TNC expression in callus results in decreased callus formation and impaired fracture healing. Conversely, administration of exogenous TNC promoted chondrogenic differentiation, cartilage template formation and ultimately improved fracture healing. Both the Hedgehog and Hippo signaling pathways were found to be involved in the pro-chondrogenic function of TNC. Our observations demonstrate that TNC is a crucial factor responsible for endochondral ossification in fracture healing and provide a potential therapeutic strategy for promoting fracture healing.


Asunto(s)
Fracturas del Fémur , Curación de Fractura , Osteogénesis , Tenascina , Animales , Masculino , Ratones , Callo Óseo/patología , Fracturas del Fémur/patología , Erizos , Vía de Señalización Hippo , Ratones Endogámicos C57BL , Tenascina/genética , Tenascina/metabolismo
11.
BMC Musculoskelet Disord ; 25(1): 180, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38413962

RESUMEN

PURPOSE: Previous studies have shown that DNA methyltransferase 3b (Dnmt3b) is the only Dnmt responsive to fracture repair and Dnmt3b ablation in Prx1-positive stem cells and chondrocyte cells both delayed fracture repair. Our study aims to explore the influence of Dnmt3b ablation in Gli1-positive stem cells in fracture healing mice and the underlying mechanism. METHODS: We generated Gli1-CreERT2; Dnmt3bflox/flox (Dnmt3bGli1ER) mice to operated tibia fracture. Fracture callus tissues of Dnmt3bGli1ER mice and control mice were collected and analyzed by X-ray, micro-CT, biomechanical testing, histopathology and TUNEL assay. RESULTS: The cartilaginous callus significantly decrease in ablation of Dnmt3b in Gli1-positive stem cells during fracture repair. The chondrogenic and osteogenic indicators (Sox9 and Runx2) in the fracture healing tissues in Dnmt3bGli1ER mice much less than control mice. Dnmt3bGli1ER mice led to delayed bone callus remodeling and decreased biomechanical properties of the newly formed bone during fracture repair. Both the expressions of Caspase-3 and Caspase-8 were upregulated in Dnmt3bGli1ER mice as well as the expressions of BCL-2. CONCLUSIONS: Our study provides an evidence that Dnmt3b ablation Gli1-positive stem cells can affect fracture healing and lead to poor fracture healing by regulating apoptosis to decrease chondrocyte hypertrophic maturation.


Asunto(s)
Callo Óseo , Fracturas de la Tibia , Animales , Ratones , Apoptosis , Callo Óseo/patología , Curación de Fractura/fisiología , Fracturas de la Tibia/cirugía , Proteína con Dedos de Zinc GLI1
12.
Curr Osteoporos Rep ; 22(1): 182-192, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38294715

RESUMEN

PURPOSE OF REVIEW: Fractures are a prominent form of traumatic injury and shall continue to be for the foreseeable future. While the inflammatory response and the cells of the bone marrow microenvironment play significant roles in fracture healing, the nervous system is also an important player in regulating bone healing. RECENT FINDINGS: Considerable evidence demonstrates a role for nervous system regulation of fracture healing in a setting of traumatic injury to the brain. Although many of the impacts of the nervous system on fracture healing are positive, pain mediated by the nervous system can have detrimental effects on mobilization and quality of life. Understanding the role the nervous system plays in fracture healing is vital to understanding fracture healing as a whole and improving quality of life post-injury. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.


Asunto(s)
Curación de Fractura , Fracturas Óseas , Humanos , Curación de Fractura/fisiología , Inteligencia Artificial , Calidad de Vida , Callo Óseo
13.
J Mech Behav Biomed Mater ; 151: 106356, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38181571

RESUMEN

In this paper, the effects of different composite plates on the healing of femoral fractures were studied by numerical simulation. The healing model of femoral fracture was established by ABAQUS display solver. Based on the fuzzy logic theory, the process of callus differentiation at femoral fracture was considered under the joint action of biological variables and mechanical stimulation, and the healing process of femur was simulated. Compare the stress on the screw, concentration of callus bone and cartilage, and callus healing performance of the carbon/epoxy composite (WSN3k) plate, glass/polypropylene composite (Twintex) plate, and stainless steel plate at various stages of bone healing, and investigate the impact of composite plates on the bone healing process. The results showed that the modulus of the plate had a notable impact on bone healing. Compared to stainless steel plate, the composite plate (due to its lower stiffness) exhibiting superior healing performance. Altering the sequence of composite laminates may modify the bone healing efficiency, and the wsn3k [0]18 composite plate exhibits superior healing performance.


Asunto(s)
Fracturas del Fémur , Acero Inoxidable , Humanos , Fracturas del Fémur/cirugía , Cicatrización de Heridas , Fémur/diagnóstico por imagen , Placas Óseas , Callo Óseo , Curación de Fractura , Fijación Interna de Fracturas/métodos , Fenómenos Biomecánicos
14.
Comput Methods Programs Biomed ; 244: 107952, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38039922

RESUMEN

BACKGROUND: Osteoporosis and diabetes are two prevalent conditions among the elderly population. Each of these conditions can profoundly influence the fracture healing process by disturbing the associated inflammatory process. However, the combined effects of osteoporosis and diabetes on fracture healing remain unclear. Therefore, the purpose of the present study is to investigate the role of osteoporosis and diabetes in fracture healing and the underlying mechanisms by developing numerical models. METHOD: This study introduces a numerical model that consists of a three-dimensional model of a tibia fracture stabilized by a Locking Compression Plate (LCP), coupled with a two-dimensional axisymmetric model which illustrates the transport and reactions of cells and cytokines throughout the inflammatory phase in early fracture healing. First, the model parameters were calibrated using available experimental data. The model was then implemented to predict the healing outcomes of fractures under five varied conditions, consisting of both osteoporotic and non-osteoporotic bones, each subjected to different physiological loads. RESULTS: The instability of the fracture callus can significantly escalate in osteoporotic fractures (e.g., when a 150 N physiological load is applied, the unstable region of the osteoporotic fracture callus can reach 26 %, in contrast to 12 % in non-osteoporotic fractures). Additionally, the mesenchymal stem cells (MSCs) proliferation and differentiation can be disrupted in osteoporotic fracture compared to non-osteoporotic fractures (e.g., on the 10th day post-fracture, the decrease in the concentration of MSCs, osteoblasts, and chondrocytes in osteoporotic fractures is nearly double that in non-osteoporotic fractures under a 150 N). Finally, the healing process of fractures can suffer significant impairment when osteoporosis coexists with diabetes (e.g., the concentration of MSCs can be drastically reduced by nearly 37 % in osteoporotic fractures under diabetic conditions when subjected to a load of 200 N) CONCLUSIONS: Fracture calluses destabilized by osteoporosis can negatively affect the fracture healing process by disrupting the proliferation and differentiation of mesenchymal stem cells (MSCs). Moreover, when osteoporosis coexists with diabetes, the fracture healing process can severely impair the fracture healing outcomes.


Asunto(s)
Diabetes Mellitus , Osteoporosis , Fracturas Osteoporóticas , Anciano , Humanos , Curación de Fractura , Osteoporosis/complicaciones , Callo Óseo
15.
Bone ; 179: 116978, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37993038

RESUMEN

Almost all prior mouse fracture healing models have used needles or K-wires for fixation, unwittingly providing inadequate mechanical stability during the healing process. Our contention is that the reported outcomes have predominantly reflected this instability, rather than the impact of diverse biological conditions, pharmacologic interventions, exogenous growth factors, or genetic considerations. This important issue becomes obvious upon a critical review of the literature. Therefore, the primary aim of this study was to demonstrate the significance of mouse-specific implants designed to provide both axial and torsional stability (Screw and IM Nail) compared to conventional pins (Needle and K-wires), even when used in mice with differently sized marrow canals and diverse genetic backgrounds. B6 (large medullary canal), DBA, and C3H (smaller medullary canals) mice were employed, all of which have different bone morphologies. Closed femoral fractures were created and stabilized with intramedullary implants that provide different mechanical conditions during the healing process. The most important finding of this study was that appropriately designed mouse-specific implants, providing both axial and torsional stability, had the greatest influence on bone healing outcomes regardless of the different bone morphologies encountered. For instance, unstable implants in the B6 strain (largest medullary canal) resulted in significantly greater callus, with a fracture region mainly comprising trabecular bone along with the presence of cartilage 28 days after surgery. The DBA and C3H strains (with smaller medullary canals) instead formed significantly less callus, and only had a small amount of intracortical trabeculation remaining. Moreover, with more stable fracture fixation a higher BV/TV was observed and cortices were largely restored to their original dimensions and structure, indicating an accelerated healing and remodeling process. These observations reveal that the diaphyseal cortical thickness, influenced by the genetic background of each strain, played a pivotal role in determining the amount of bone formation in response to the fracture. These findings are highly important, indicating the rate and type of tissue formed is a direct result of mechanical instability, and this most likely would mask the true contribution of the tested genes, genetic backgrounds, or various therapeutic agents administered during the bone healing process.


Asunto(s)
Fracturas del Fémur , Fijación Intramedular de Fracturas , Ratones , Animales , Curación de Fractura/fisiología , Ratones Endogámicos DBA , Ratones Endogámicos C3H , Fracturas del Fémur/cirugía , Callo Óseo
16.
Bone ; 179: 116983, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38013019

RESUMEN

Stress fractures occur as a result of repeated mechanical stress on bone and are commonly found in the load-bearing lower extremities. Macrophages are key players in the immune system and play an important role in bone remodeling and fracture healing. However, the role of macrophages in stress fractures has not been adequately addressed. We hypothesize that macrophage infiltration into a stress fracture callus site promotes bone healing. To test this, a unilateral stress fracture induction model was employed in which the murine ulna of four-month-old, C57BL/6 J male mice was repeatedly loaded with a pre-determined force until the bone was displaced a distance below the threshold for complete fracture. Mice were treated daily with parathyroid hormone (PTH, 50 µg/kg/day) starting two days before injury and continued until 24 h before euthanasia either four or six days after injury, or treated with trabectedin (0.15 mg/kg) on the day of stress fracture and euthanized three or seven days after injury. These treatments were used due to their established effects on macrophages. While macrophages have been implicated in the anabolic effects of PTH, trabectedin, an FDA approved chemotherapeutic, compromises macrophage function and reduces bone mass. At three- and four-days post injury, callus macrophage numbers were analyzed histologically. There was a significant increase in macrophages with PTH treatment compared to vehicle in the callus site. By one week of healing, treatments differentially affected the bony callus as analyzed by microcomputed tomography. PTH enhanced callus bone volume. Conversely, callus bone volume was decreased with trabectedin treatment. Interestingly, concurrent treatment with PTH and trabectedin rescued the reduction observed in the callus with trabectedin treatment alone. This study reports on the key involvement of macrophages during stress fracture healing. Given these observed outcomes on macrophage physiology and bone healing, these findings may be important for patients actively receiving either of these FDA-approved therapeutics.


Asunto(s)
Fracturas por Estrés , Hormona Paratiroidea , Humanos , Masculino , Ratones , Animales , Lactante , Hormona Paratiroidea/farmacología , Hormona Paratiroidea/uso terapéutico , Trabectedina/farmacología , Fracturas por Estrés/tratamiento farmacológico , Fracturas por Estrés/patología , Microtomografía por Rayos X/métodos , Ratones Endogámicos C57BL , Callo Óseo/patología , Curación de Fractura , Macrófagos
17.
Jt Dis Relat Surg ; 35(1): 146-155, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38108176

RESUMEN

OBJECTIVES: The purpose of this study was to investigate whether hydroxychloroquine (HCQ) sulfate causes oxidative stress (OS) and its effect on fracture healing in an experimental rat model. MATERIALS AND METHODS: In this experimental study, open diaphyseal femur fractures were induced in 24 eight-week-old male rats (mean weight: 225±25 g; range, 200 to 250 g) and then fixed with K-wire. The rats were divided into four groups: HCQ-2, control-2 (C-2), HCQ-4, and control-4 (C-4). During the study period, rats in the HCQ groups received an HCQ solution (160 mg/kg/day), whereas rats in the control groups received saline. The HCQ-2 and C-2 groups were sacrificed on the 14th day, and the HCQ-4 and C-4 groups were sacrificed on the 28th day. After sacrifice, malondialdehyde levels induced by OS were calculated for each rat, and fracture healing was evaluated radiographically, histomorphometrically, histopathologically, and immunohistochemically. RESULTS: Malondialdehyde levels were higher in the HCQ groups than in the control groups (p<0.05). Hydroxychloroquine caused OS in rats. The ratio of total callus diameter to femur bone diameter was lower in HCQ groups compared to control groups (p<0.05). No differences were observed when comparing radiological and histological healing results between the control and HCQ groups. Alkaline phosphatase levels were lower in the HCQ-4 group than the C-4 group at week four (p<0.05), although osteocalcin and osteopontin levels did not differ between groups (p>0.05). Oxidative stress had no adverse effects on histologic healing outcomes and osteoblast functions. Cathepsin K and tartrate-resistant acid phosphatase-5b levels were higher in the HCQ-4 group than in the C-4 group (p<0.05). While the number and function of osteoclasts increased due to OS in callus tissue, a decrease in the number of chondrocytes was observed. CONCLUSION: Hydroxychloroquine-induced OS increases the number and function of osteoclasts and decreases the number of hypertrophic chondrocytes and endochondral ossification but has no significant effect on mid-late osteoblast products and histological fracture healing scores.


Asunto(s)
Fracturas del Fémur , Curación de Fractura , Ratas , Masculino , Animales , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Callo Óseo , Osteogénesis , Fracturas del Fémur/inducido químicamente , Fracturas del Fémur/diagnóstico por imagen , Fracturas del Fémur/tratamiento farmacológico , Malondialdehído/farmacología
18.
Elife ; 122023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-38079220

RESUMEN

Insufficient bone fracture repair represents a major clinical and societal burden and novel strategies are needed to address it. Our data reveal that the transforming growth factor-ß superfamily member Activin A became very abundant during mouse and human bone fracture healing but was minimally detectable in intact bones. Single-cell RNA-sequencing revealed that the Activin A-encoding gene Inhba was highly expressed in a unique, highly proliferative progenitor cell (PPC) population with a myofibroblast character that quickly emerged after fracture and represented the center of a developmental trajectory bifurcation producing cartilage and bone cells within callus. Systemic administration of neutralizing Activin A antibody inhibited bone healing. In contrast, a single recombinant Activin A implantation at fracture site in young and aged mice boosted: PPC numbers; phosphorylated SMAD2 signaling levels; and bone repair and mechanical properties in endochondral and intramembranous healing models. Activin A directly stimulated myofibroblastic differentiation, chondrogenesis and osteogenesis in periosteal mesenchymal progenitor culture. Our data identify a distinct population of Activin A-expressing PPCs central to fracture healing and establish Activin A as a potential new therapeutic tool.


Asunto(s)
Activinas , Callo Óseo , Curación de Fractura , Ratones , Humanos , Animales , Curación de Fractura/genética , Osteogénesis , Células Madre , Diferenciación Celular
19.
Sensors (Basel) ; 23(23)2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-38067749

RESUMEN

Calluses are thickened skin areas that develop due to repeated friction, pressure, or other types of irritation. While calluses are usually harmless and formed as a protective surface, they can lead to skin ulceration or infection if left untreated. As calluses are often not clearly visible to the patients, and some areas of dead skin can be missed during debridement, accessory tools can be useful in assessment and follow-up. The practical question addressed in this article is whether or not thermal imaging adds value to callus assessment. We have performed a theoretical analysis of the feasibility of thermographic imaging for callus identification. Our analytical calculations show that the temperature drop in the epidermis should be on the order of 0.1 °C for the normal epidermis in hairy skin, 0.9 °C for glabrous skin, and 1.5-2 °C or higher in calluses. We have validated our predictions on gelatin phantoms and demonstrated the feasibility of thermographic imaging for callus identification in two clinical case series. Our experimental results are in agreement with theoretical predictions and support the notion that local skin temperature variations can indicate epidermis thickness variations, which can be used for callus identification. In particular, a surface temperature drop on the order of 0.5 °C or more can be indicative of callus presence, particularly in callus-prone areas. In addition, our analytical calculations and phantom experiments show the importance of ambient temperature measurements during thermographic assessments.


Asunto(s)
Callosidades , Humanos , Termografía/métodos , Piel , Callo Óseo , Temperatura Cutánea
20.
PLoS One ; 18(12): e0296510, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38157369

RESUMEN

INTRODUCTION: Tissue engineering has emerged as an innovative approach to treat critical-size bone defects using biocompatible scaffolds, thus avoiding complex distraction surgeries or limited stock grafts. Continuous regeneration monitoring is essential in critical-size cases due to the frequent appearance of non-unions. This work evaluates the potential clinical use of gait analysis for the mechanical assessment of a tissue engineering regeneration as an alternative to the traditional and hardly conclusive manual or radiological follow-up. MATERIALS AND METHODS: The 15-mm metatarsal fragment of eight female merino sheep was surgically replaced by a bioceramic scaffold stabilized with an external fixator. Gait tests were performed weekly by making the sheep walk on an instrumented gangway. The evolution of different kinematic and dynamic parameters was analyzed for all the animal's limbs, as well as asymmetries between limbs. Finally, potential correlation in the recovery of the gait parameters was evaluated through the linear regression models. RESULTS: After surgery, the operated limb has an altered way of carrying body weight while walking. Its loading capacity was significantly reduced as the stance phases were shorter and less impulsive. The non-operated limbs compensated for this mobility deficit. All parameters were normalizing during the consolidation phase while the bone callus was simultaneously mineralizing. The results also showed high levels of asymmetry between the operated limb and its contralateral, which exceeded 150% when analyzing the impulse after surgery. Gait recovery significantly correlated between symmetrical limbs. CONCLUSIONS: Gait analysis was presented as an effective, low-cost tool capable of mechanically predicting the regeneration of critical-size defects treated by tissue engineering, as comparing regeneration processes or novel scaffolds. Despite the progressive normalization as the callus mineralized, the bearing capacity reduction and the asymmetry of the operated limb were more significant than in other orthopedic alternatives.


Asunto(s)
Regeneración Ósea , Ingeniería de Tejidos , Femenino , Ovinos , Animales , Marcha , Callo Óseo , Caminata , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA