Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
1.
Biomater Adv ; 159: 213805, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38457904

RESUMO

Bone defects may occur in different sizes and shapes due to trauma, infections, and cancer resection. Autografts are still considered the primary treatment choice for bone regeneration. However, they are hard to source and often create donor-site morbidity. Injectable microgels have attracted much attention in tissue engineering and regenerative medicine due to their ability to replace inert implants with a minimally invasive delivery. Here, we developed novel cell-laden bioprinted gelatin methacrylate (GelMA) injectable microgels, with controllable shapes and sizes that can be controllably mineralized on the nanoscale, while stimulating the response of cells embedded within the matrix. The injectable microgels were mineralized using a calcium and phosphate-rich medium that resulted in nanoscale crystalline hydroxyapatite deposition and increased stiffness within the crosslinked matrix of bioprinted GelMA microparticles. Next, we studied the effect of mineralization in osteocytes, a key bone homeostasis regulator. Viability stains showed that osteocytes were maintained at 98 % viability after mineralization with elevated expression of sclerostin in mineralized compared to non-mineralized microgels, showing that mineralization can effectively enhances osteocyte maturation. Based on our findings, bioprinted mineralized GelMA microgels appear to be an efficient material to approximate the bone microarchitecture and composition with desirable control of sample injectability and polymerization. These bone-like bioprinted mineralized biomaterials are exciting platforms for potential minimally invasive translational methods in bone regenerative therapies.


Assuntos
Gelatina , Microgéis , Gelatina/farmacologia , Gelatina/química , Materiais Biocompatíveis , Metacrilatos/química
2.
ACS Biomater Sci Eng ; 9(11): 6282-6292, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37906515

RESUMO

In order to scale up culture therapeutic cells, such as mesenchymal stromal cells (MSCs), culture in suspension bioreactors using microcarriers (µCs) is preferred. However, the impact of microcarrier type on the resulting MSC secretory activity has not been investigated. In this study, two poly(ethylene glycol) hydrogel formulations with different swelling ratios (named "stiffer" and "softer") were fabricated as µC substrates to culture MSCs and MSCs genetically modified to express the interleukin-1 receptor antagonist (IL-1Ra-MSCs). Changes in cell number, secretory and angiogenic activity, and changes in MAPK signaling were evaluated when cultured on hydrogel µCs, as well as on tissue culture plastic-based Synthemax µCs. We demonstrated that culture on stiffer µCs increased secretion of IL-1Ra compared to culture on Synthemax µCs by IL-1Ra-MSCs by 1.2- to 1.6-fold, as well as their in vitro angiogenic activity, compared to culture on Synthemax µCs, while culture on both stiffer and softer µCs altered the secretion of several other factors compared to culture on Synthemax µCs. Changes in angiogenic activity corresponded with increased gene expression and secretion of hepatocyte growth factor by MSCs cultured on softer µCs by 2.5- to 6-fold compared to MSCs cultured on Synthemax µCs. Quantification of phosphoprotein signaling with the MAPK pathway revealed broad reduction of pathway activation by IL-1Ra-MSCs cultured on both stiffer and softer µCs compared to Synthemax, where phosphorylated c-Jun, ATF2, and MEK1 were reduced specifically on softer µCs. Overall, this study showed that µC surfaces can influence the secretory activity of genetically modified MSCs and identified associated changes in MAPK pathway signaling, which is a known central regulator of cytokine secretion.


Assuntos
Proteína Antagonista do Receptor de Interleucina 1 , Células-Tronco Mesenquimais , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Células-Tronco Mesenquimais/metabolismo , Técnicas de Cultura de Células/métodos , Materiais Biocompatíveis , Hidrogéis/farmacologia , Hidrogéis/metabolismo , Polietilenoglicóis/farmacologia , Polietilenoglicóis/metabolismo
3.
Res Sq ; 2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37886569

RESUMO

Mechanical loading is integral to bone development and repair. The application of mechanical loads through rehabilitation are regularly prescribed as a clinical aide following severe bone injuries. However, current rehabilitation regimens typically involve long periods of non-loading and rely on subjective patient feedback, leading to muscle atrophy and soft tissue fibrosis. While many pre-clinical studies have focused on unloading, ambulatory loading, or direct mechanical compression, rehabilitation intensity and its impact on the local strain environment and subsequent bone healing have largely not been investigated. This study combines implantable strain sensors and subject-specific finite element models in a pre-clinical rodent model with a defect size on the cusp of critically-sized. Animals were enrolled in either high or low intensity rehabilitation one week post injury to investigate how rehabilitation intensity affects the local mechanical environment and subsequent functional bone regeneration. The high intensity rehabilitation animals were given free access to running wheels with resistance, which increased local strains within the regenerative niche by an average of 44% compared to the low intensity (no-resistance) group. Finite element modeling demonstrated that resistance rehabilitation significantly increased compressive strain by a factor of 2.0 at week 1 and 4.45 after 4 weeks of rehabilitation. The resistance rehabilitation group had significantly increased regenerated bone volume and higher bone bridging rates than its sedentary counterpart (bone volume: 22.00 mm3 ± 4.26 resistance rehabilitation vs 8.00 mm3 ± 2.27 sedentary; bridging rates: 90% resistance rehabilitation vs 50% sedentary). In addition, animals that underwent resistance running had femurs with improved mechanical properties compared to those left in sedentary conditions, with failure torque and torsional stiffness values matching their contralateral, intact femurs (stiffness: 0.036 Nm/deg ± 0.006 resistance rehabilitation vs 0.008 Nm/deg ± 0.006 sedentary). Running on a wheel with no resistance rehabilitation also increased bridging rates (100% no resistance rehabilitation vs 50% sedentary). Analysis of bone volume and von Frey suggest no-resistance rehabilitation may improve bone regeneration and hindlimb functionality. These results demonstrate the potential for early resistance rehabilitation as a rehabilitation regimen to improve bone regeneration and functional recovery.

4.
bioRxiv ; 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37873385

RESUMO

Bone defects may occur in different sizes and shapes due to trauma, infections, and cancer resection. Autografts are still considered the primary treatment choice for bone regeneration. However, they are hard to source and often create donor-site morbidity. Injectable microgels have attracted much attention in tissue engineering and regenerative medicine due to their ability to replace inert implants with a minimally invasive delivery. Here, we developed novel cell-laden bioprinted gelatin methacrylate (GelMA) injectable microgels, with controllable shapes and sizes that can be controllably mineralized on the nanoscale, while stimulating the response of cells embedded within the matrix. The injectable microgels were mineralized using a calcium and phosphate-rich medium that resulted in nanoscale crystalline hydroxyapatite deposition and increased stiffness within the crosslinked matrix of bioprinted GelMA microparticles. Next, we studied the effect of mineralization in osteocytes, a key bone homeostasis regulator. Viability stains showed that osteocytes were maintained at 98% viability after mineralization with elevated expression of sclerostin in mineralized compared to non-mineralized microgels, indicating that mineralization effectively enhances osteocyte maturation. Based on our findings, bioprinted mineralized GelMA microgels appear to be an efficient material to approximate the bone microarchitecture and composition with desirable control of sample injectability and polymerization. These bone-like bioprinted mineralized biomaterials are exciting platforms for potential minimally invasive translational methods in bone regenerative therapies.

5.
Front Bioeng Biotechnol ; 11: 1224141, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37744252

RESUMO

Background: Micronized dehydrated human amnion/chorion membrane (mdHACM) has reduced short term post-traumatic osteoarthritis (PTOA) progression in rats when delivered 24 h after medial meniscal transection (MMT) and is being investigated for clinical use as a disease modifying therapy. Much remains to be assessed, including its potential for longer-term therapeutic benefit and treatment effects after onset of joint degeneration. Objectives: Characterize longer-term effects of acute treatment with mdHACM and determine whether treatment administered to joints with established PTOA could slow or reverse degeneration. Hypotheses: Acute treatment effects will be sustained for 6 weeks, and delivery of mdHACM after onset of joint degeneration will attenuate structural osteoarthritic changes. Methods: Rats underwent MMT or sham surgery (left leg). mdHACM was delivered intra-articularly 24 h or 3 weeks post-surgery (n = 5-7 per group). Six weeks post-surgery, animals were euthanized and left tibiae scanned using equilibrium partitioning of an ionic contrast agent microcomputed tomography (EPIC-µCT) to structurally quantify joint degeneration. Histology was performed to examine tibial plateau cartilage. Results: Quantitative 3D µCT showed that cartilage structural metrics (thickness, X-ray attenuation, surface roughness, exposed bone area) for delayed mdHACM treatment limbs were significantly improved over saline treatment and not significantly different from shams. Subchondral bone mineral density and thickness for the delayed treatment group were significantly improved over acute treated, and subchondral bone thickness was not significantly different from sham. Marginal osteophyte degenerative changes were decreased with delayed mdHACM treatment compared to saline. Acute treatment (24 h post-surgery) did not reduce longer-term joint tissue degeneration compared to saline. Histology supported µCT findings and further revealed that while delayed treatment reduced cartilage damage, chondrocytes displayed qualitatively different morphologies and density compared to sham. Conclusion: This study provides insight into effects of intra-articular delivery timing relative to PTOA progression and the duration of therapeutic benefit of mdHACM. Results suggest that mdHACM injection into already osteoarthritic joints can improve joint health, but a single, acute mdHACM injection post-injury does not prevent long term osteoarthritis associated with meniscal instability. Further work is needed to fully characterize the durability of therapeutic benefit in stable osteoarthritic joints and the effects of repeated injections.

6.
Sci Transl Med ; 15(707): eabp8258, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37531418

RESUMO

Translational impact assessment is key to selecting those biomedical research discoveries most likely to be converted into viable new products to improve human health. However, metrics for translational success are variable, are not limited to commercial success, and may not be relevant to every case or institution. Societal impact is a top translational priority in a globalized society.


Assuntos
Pesquisa Biomédica , Pesquisa Translacional Biomédica , Humanos , Benchmarking
7.
Stem Cells Transl Med ; 12(5): 293-306, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37184892

RESUMO

Juvenile osteochondritis dissecans (JOCD) is a pediatric disease, which begins with an osteonecrotic lesion in the secondary ossification center which, over time, results in the separation of the necrotic fragment from the parent bone. JOCD predisposes to early-onset osteoarthritis. However, the knowledge gap in JOCD pathomechanisms severely limits current therapeutic strategies. To elucidate its etiology, we conducted a study with induced pluripotent stem cells (iPSCs) from JOCD and control patients. iPSCs from skin biopsies were differentiated to iMSCs (iPSC-derived mesenchymal stromal cells) and subjected to chondrogenic and endochondral ossification, and endoplasmic reticulum (ER)-stress induction assays. Our study, using 3 JOCD donors, showed that JOCD cells have lower chondrogenic capability and their endochondral ossification process differs from control cells; yet, JOCD- and control-cells accomplish osteogenesis of similar quality. Our findings show that endoplasmic reticulum stress sensing and response mechanisms in JOCD cells, which partially regulate chondrocyte and osteoblast differentiation, are related to these differences. We suggest that JOCD cells are more sensitive to ER stress than control cells, and in pathological microenvironments, such as microtrauma and micro-ischemia, JOCD pathogenesis pathways may be initiated. This study is the first, to the best of our knowledge, to realize the important role that resident cells and their differentiating counterparts play in JOCD and to put forth a novel etiological hypothesis that seeks to consolidate and explain previously postulated hypotheses. Furthermore, our results establish well-characterized JOCD-specific iPSC-derived in vitro models and identified potential targets which could be used to improve diagnostic tools and therapeutic strategies in JOCD.


Assuntos
Células-Tronco Pluripotentes Induzidas , Osteocondrite Dissecante , Criança , Humanos , Osteocondrite Dissecante/diagnóstico , Osteocondrite Dissecante/patologia , Osteocondrite Dissecante/terapia , Células-Tronco Pluripotentes Induzidas/patologia , Necrose/complicações , Condrócitos , Estresse do Retículo Endoplasmático
8.
Sensors (Basel) ; 23(4)2023 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-36850431

RESUMO

Magnetoelastic sensors, which undergo mechanical resonance when interrogated with magnetic fields, can be functionalized to measure various physical quantities and chemical/biological analytes by tracking their resonance behaviors. The unique wireless and functionalizable nature of these sensors makes them good candidates for biological sensing applications, from the detection of specific bacteria to tracking force loading inside the human body. In this study, we evaluate the viability of magnetoelastic sensors based on a commercially available magnetoelastic material (Metglas 2826 MB) for wirelessly monitoring the attachment and growth of human mesenchymal stromal cells (hMSCs) in 2D in vitro cell culture. The results indicate that the changes in sensor resonance are linearly correlated with cell quantity. Experiments using a custom-built monitoring system also demonstrated the ability of this technology to collect temporal profiles of cell growth, which could elucidate key stages of cell proliferation based on acute features in the profile. Additionally, there was no observed change in the morphology of cells after they were subjected to magnetic and mechanical stimuli from the monitoring system, indicating that this method for tracking cell growth may have minimal impact on cell quality and potency.


Assuntos
Células-Tronco Mesenquimais , Humanos , Proliferação de Células , Técnicas de Cultura de Células , Cultura , Campos Magnéticos
9.
J Biomed Mater Res A ; 111(5): 634-643, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36794576

RESUMO

Stimuli-responsive biomaterials may be used to better control the release of bioactive molecules or cells for applications involving drug delivery and controlled cell release. In this study, we developed a Factor Xa (FXa)-responsive biomaterial capable of controlled release of pharmaceutical agents and cells from in vitro culture. FXa-cleavable substrates were formed as hydrogels that degraded in response to FXa enzyme over several hours. Hydrogels were shown to release both heparin and a model protein in response to FXa. Additionally, RGD-functionalized FXa-degradable hydrogels were used to culture mesenchymal stromal cells (MSCs), enabling FXa-mediated cell dissociation from hydrogels in a manner that preserved multicellular structures. Harvesting MSCs using FXa-mediated dissociation did not influence their differentiation capacity or indoleamine 2,3-dioxygenase (IDO) activity (a measure of immunomodulatory capacity). In all, this FXa-degradable hydrogel is a novel responsive biomaterial system that may be used for on-demand drug delivery, as well as for improving processes for in vitro culture of therapeutic cells.


Assuntos
Produtos Biológicos , Fator Xa , Hidrogéis/química , Materiais Biocompatíveis/química , Técnicas de Cultura de Células
10.
Adv Healthc Mater ; 12(11): e2200976, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36808718

RESUMO

Bone autografts remain the gold standard for bone grafting surgeries despite having increased donor site morbidity and limited availability. Bone morphogenetic protein-loaded grafts represent another successful commercial alternative. However, the therapeutic use of recombinant growth factors has been associated with significant adverse clinical outcomes. This highlights the need to develop biomaterials that closely approximate the structure and composition of bone autografts, which are inherently osteoinductive and biologically active with embedded living cells, without the need for added supplements. Here, injectable growth factor-free bone-like tissue constructs are developed, that closely approximate the cellular, structural, and chemical composition of bone autografts. It is demonstrated that these micro-constructs are inherently osteogenic, and demonstrate the ability to stimulate mineralized tissue formation and regenerate bone in critical-sized defects in-vivo. Furthermore, the mechanisms that allow human mesenchymal stem cells (hMSCs) to be highly osteogenic in these constructs, despite the lack of osteoinductive supplements, are assessed, whereby Yes activated protein (YAP) nuclear localization and adenosine signaling appear to regulate osteogenic cell differentiation. The findings represent a step toward a new class of minimally invasive, injectable, and inherently osteoinductive scaffolds, which are regenerative by virtue of their ability to mimic the tissue cellular and extracellular microenvironment, thus showing promise for clinical applications in regenerative engineering.


Assuntos
Microgéis , Humanos , Regeneração Óssea/fisiologia , Osteogênese/fisiologia , Osso e Ossos , Materiais Biocompatíveis/química , Diferenciação Celular/fisiologia , Engenharia Tecidual , Alicerces Teciduais/química
11.
Biomed Phys Eng Express ; 9(2)2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36716480

RESUMO

Fluid shear stress (FSS) is an important parameter that regulates various cell functions such as proliferation and migration. While there are a number of techniques to generate FSSin vitro, many of them require physical deformation or movement of solid objects to generate the fluid shear, making it difficult to decouple the effects of FSS and mechanical strains. This work describes the development of a non-mechanical means to generate fluid flow and FSS in a 2Din vitrosetting. This was accomplished with a magnetohydrodynamic (MHD) pump, which creates liquid flow by generating a Lorentz force through the interaction between an electric field and an orthogonal magnetic field. The MHD pump system presented here consisted of trapezoidal prism-shaped magnets, a pair of platinum electrodes, and a modified petri dish. The system was validated and tested on anin vitrowound model, which is based on analyzing the migration of fibroblast cells through an artificially created scratch on a confluent cell culture surface. Experiments were performed to a control group, an electric field only group, and a group that was subject to fluid flow with the application of both electric field and magnetic field. Results show that fibroblast cells that experienced fluid shear have higher wound closure rate compared to the control group and the electric field only group. The data shows that the MHD pump can be a great tool to study FSSin vitro. Furthermore, due to its fluid flow generation without mechanical force, this system can be adapted and implemented to study the role of FSS and electric field on wound healingin vivo.


Assuntos
Movimento Celular , Estresse Mecânico
12.
J Orthop Res ; 41(1): 130-140, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35340049

RESUMO

Treatment of large bone defects with supraphysiological doses of bone morphogenetic protein-2 (BMP-2) has been associated with complications including heterotopic ossification (HO), inflammation, and pain, presumably due to poor spatiotemporal control of BMP-2. We have previously recapitulated extensive HO in our rat femoral segmental defect model by treatment with high-dose BMP-2 (30 µg). Using this model and BMP-2 dose, our objective was to evaluate the utility of a clinically available human amniotic membrane (AM) around the defect space for guided bone regeneration and reduction of HO. We hypothesized that AM surrounding collagen sponge would attenuate heterotopic ossification compared with collagen sponge alone. In vitro, AM retained more BMP-2 than a synthetic poly(ε-caprolactone) membrane through 21 days. In vivo, as hypothesized, the collagen + AM resulted in significantly less heterotopic ossification and correspondingly, lower total bone volume (BV), compared with collagen sponge alone. Although bone formation within the defect was delayed with AM around the defect, by 12 weeks, defect BVs were equivalent. Torsional stiffness was significantly reduced with AM but was equivalent to that of intact bone. Collagen + AM resulted in the formation of dense fibrous tissue and mineralized tissue, while the collagen group contained primarily mineralized tissue surrounded by marrow-like structures. Especially in conjunction with high doses of growth factor delivered via collagen sponge, these findings suggest AM may be effective as an overlay adjacent to bone healing sites to spatially direct bone regeneration and minimize heterotopic ossification.


Assuntos
Âmnio , Colágeno , Humanos , Animais , Ratos , Proteínas Morfogenéticas Ósseas
13.
Sci Transl Med ; 14(666): eabn9074, 2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36223445

RESUMO

Musculoskeletal injuries and disorders are the leading cause of physical disability worldwide and a considerable socioeconomic burden. The lack of effective therapies has driven the development of novel bioengineering approaches that have recently started to gain clinical approvals. In this review, we first discuss the self-repair capacity of the musculoskeletal tissues and describe causes of musculoskeletal dysfunction. We then review the development of novel biomaterial, immunomodulatory, cellular, and gene therapies to treat musculoskeletal disorders. Last, we consider the recent regulatory changes and future areas of technological progress that can accelerate translation of these therapies to clinical practice.


Assuntos
Materiais Biocompatíveis , Bioengenharia , Engenharia Biomédica , Terapia Genética , Engenharia Tecidual , Cicatrização
14.
Front Surg ; 9: 934773, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35874126

RESUMO

Bone non-unions resulting from severe traumatic injuries pose significant clinical challenges, and the biological factors that drive progression towards and healing from these injuries are still not well understood. Recently, a dysregulated systemic immune response following musculoskeletal trauma has been identified as a contributing factor for poor outcomes and complications such as infections. In particular, myeloid-derived suppressor cells (MDSCs), immunosuppressive myeloid-lineage cells that expand in response to traumatic injury, have been highlighted as a potential therapeutic target to restore systemic immune homeostasis and ultimately improve functional bone regeneration. Previously, we have developed a novel immunomodulatory therapeutic strategy to deplete MDSCs using Janus gold nanoparticles that mimic the structure and function of antibodies. Here, in a preclinical delayed treatment composite injury model of bone and muscle trauma, we investigate the effects of these nanoparticles on circulating MDSCs, systemic immune profiles, and functional bone regeneration. Unexpectedly, treatment with the nanoparticles resulted in depletion of the high side scatter subset of MDSCs and an increase in the low side scatter subset of MDSCs, resulting in an overall increase in total MDSCs. This overall increase correlated with a decrease in bone volume (P = 0.057) at 6 weeks post-treatment and a significant decrease in mechanical strength at 12 weeks post-treatment compared to untreated rats. Furthermore, MDSCs correlated negatively with endpoint bone healing at multiple timepoints. Single cell RNA sequencing of circulating immune cells revealed differing gene expression of the SNAb target molecule S100A8/A9 in MDSC sub-populations, highlighting a potential need for more targeted approaches to MDSC immunomodulatory treatment following trauma. These results provide further insights on the role of systemic immune dysregulation for severe trauma outcomes in the case of non-unions and composite injuries and suggest the need for additional studies on targeted immunomodulatory interventions to enhance healing.

15.
Connect Tissue Res ; 63(3): 228-242, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35172654

RESUMO

PURPOSE: A healthy musculoskeletal system requires complex functional integration of bone, muscle, cartilage, and connective tissues responsible for bodily support, motion, and the protection of vital organs. Conditions or injuries to musculoskeeltal tissues can devastate an individual's quality of life. Some conditions that are particularly disabling include severe bone and muscle injuries to the extremities and amputations resulting from unmanageable musculoskeletal conditions or injuries. Monitoring and managing musculoskeletal health is intricate because of the complex mechanobiology of these interconnected tissues. METHODS: For this article, we reviewed literature on implantable biosensors related to clinical data of the musculoskeletal system, therapeutics for complex bone injuries, and osseointegrated prosthetics as example applications. RESULTS: As a result, a brief summary of biosensors technologies is provided along with review of noteworthy biosensors and future developments needed to fully realize the translational benefit of biosensors for musculoskeletal health. CONCLUSIONS: Novel implantable biosensors capable of tracking biophysical parameters in vivo are highly relevant to musculoskeletal health because of their ability to collect clinical data relevant to medical decisions, complex trauma treatment, and the performance of osseointegrated prostheses.


Assuntos
Técnicas Biossensoriais , Qualidade de Vida , Osso e Ossos , Osseointegração , Próteses e Implantes
16.
Sensors (Basel) ; 22(3)2022 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-35161572

RESUMO

Magnetoelastic sensors, typically made of magnetostrictive and magnetically-soft materials, can be fabricated from commercially available materials into a variety of shapes and sizes for their intended applications. Since these sensors are wirelessly interrogated via magnetic fields, they are good candidates for use in both research and industry, where detection of environmental parameters in closed and controlled systems is necessary. Common applications for these sensors include the investigation of physical, chemical, and biological parameters based on changes in mass loading at the sensor surface which affect the sensor's behavior at resonance. To improve the performance of these sensors, optimization of sensor geometry, size, and detection conditions are critical to increasing their mass sensitivity and detectible range. This work focuses on investigating how the geometry of the sensor influences its resonance spectrum, including the sensor's shape, size, and aspect ratio. In addition to these factors, heterogeneity in resonance magnitude was mapped for the sensor surface and the effect of the magnetic bias field strength on the resonance spectrum was investigated. Analysis of the results indicates that the shape of the sensor has a strong influence on the emergent resonant modes. Reducing the size of the sensor decreased the sensor's magnitude of resonance. The aspect ratio of the sensor, along with the bias field strength, was also observed to affect the magnitude of the signal; over or under biasing and aspect ratio extremes were observed to decrease the magnitude of resonance, indicating that these parameters can be optimized for a given shape and size of magnetoelastic sensor.


Assuntos
Técnicas Biossensoriais
17.
Connect Tissue Res ; 63(1): 16-27, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33820456

RESUMO

Purpose: Mechanical loading of bone defects through rehabilitation is a promising approach to stimulate repair and reduce nonunion risk; however, little is known about how therapeutic mechanical stimuli modulate early-stage repair before mineralized bone formation. The objective of this study was to investigate the early effects of osteogenic loading on cytokine expression and angiogenesis during the first 3 weeks of BMP-2 mediated segmental bone defect repair.Materials and Methods: A rat model of BMP-2 mediated bone defect repair was subjected to an osteogenic mechanical loading protocol using ambulatory rehabilitation and a compliant, load-sharing fixator with an integrated implantable strain sensor. The effect of fixator load-sharing on local tissue strain, angiogenesis, and cytokine expression was evaluated.Results: Using sensor readings for local measurements of boundary conditions, finite element simulations showed strain became amplified in remaining soft tissue regions between 1 and 3 weeks (Week 3: load-sharing: -1.89 ± 0.35% and load-shielded: -1.38 ± 0.35% vs. Week 1: load-sharing: -1.54 ± 0.17%; load-shielded: -0.76 ± 0.06%). Multivariate analysis of cytokine arrays revealed that load-sharing significantly altered expression profiles in the defect tissue at 2 weeks compared to load-shielded defects. Specifically, loading reduced VEGF (p = 0.052) and increased CXCL5 (LIX) levels. Subsequently, vascular volume in loaded defects was reduced relative to load-shielded defects but similar to intact bone at 3 weeks. Endochondral bone repair was also observed histologically in loaded defects at 3 weeks.Conclusions: Together, these results demonstrate that moderate ambulatory strains previously shown to stimulate bone regeneration significantly alter early angiogenic and cytokine signaling and may promote endochondral ossification.


Assuntos
Proteína Morfogenética Óssea 2 , Osteogênese , Animais , Regeneração Óssea/fisiologia , Osteogênese/fisiologia , Próteses e Implantes , Ratos
18.
Acta Biomater ; 127: 180-192, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33823326

RESUMO

Successful bone healing in severe trauma depends on early revascularization to restore oxygen, nutrient, growth factor, and progenitor cell supply to the injury. Therapeutic angiogenesis strategies have therefore been investigated to promote revascularization following severe bone injuries; however, results have been inconsistent. This is the first study investigating the effects of dual angiogenic growth factors (VEGF and PDGF) with low-dose bone morphogenetic protein-2 (BMP-2; 2.5 µg) on bone healing in a clinically challenging composite bone-muscle injury model. Our hydrogel-based delivery systems demonstrated a more than 90% protein entrapment efficiency and a controlled simultaneous release of three growth factors over 28 days. Co-stimulation of microvascular fragment constructs with VEGF and PDGF promoted vascular network formation in vitro compared to VEGF or PDGF alone. In an in vivo model of segmental bone and volumetric muscle loss injury, combined VEGF (5 µg) and PDGF (7.5 µg or 15 µg) delivery with a low dose of BMP-2 significantly enhanced regeneration of vascularized bone compared to BMP-2 treatment alone. Notably, the regenerated bone mechanics reached ~60% of intact bone, a value that was previously only achieved by delivery of high-dose BMP-2 (10 µg) in this injury model. Overall, sustained delivery of VEGF, PDFG, and BMP-2 is a promising strategy to promote functional vascularized bone tissue regeneration following severe composite musculoskeletal injury. Although this study is conducted in a clinically relevant composite injury model in rats using a simultaneous release strategy, future studies are necessary to test the regenerative potential of spatiotemporally controlled delivery of triple growth factors on bone healing using large animal models. STATEMENT OF SIGNIFICANCE: Volumetric muscle loss combined with delayed union or non-union bone defect causes deleterious effects on bone regeneration even with the supplementation of bone morphogenetic protein-2 (BMP-2). In this study, the controlled delivery of dual angiogenic growth factors (vascular endothelial growth factor [VEGF] + Platelet-derived growth factor [PDGF]) increases vascular growth in vitro. Co-delivering VEGF+PDGF significantly increase the bone formation efficacy of low-dose BMP-2 and improves the mechanics of regenerated bone in a challenging composite bone-muscle injury model.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Regeneração Óssea , Sistema Musculoesquelético/lesões , Animais , Osso e Ossos , Hidrogéis/farmacologia , Osteogênese , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ratos , Fator A de Crescimento do Endotélio Vascular/farmacologia
19.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33597299

RESUMO

Severe traumatic injuries are a widespread and challenging clinical problem, and yet the factors that drive successful healing and restoration of function are still not well understood. One recently identified risk factor for poor healing outcomes is a dysregulated immune response following injury. In a preclinical model of orthopedic trauma, we demonstrate that distinct systemic immune profiles are correlated with impaired bone regeneration. Most notably, elevated blood levels of myeloid-derived suppressor cells (MDSCs) and the immunosuppressive cytokine interleukin-10 (IL-10) are negatively correlated with functional bone regeneration as early as 1 wk posttreatment. Nonlinear multivariate regression also implicated these two factors as the most influential in predictive computational models. These results support a significant relationship between early systemic immune responses to trauma and subsequent local bone regeneration and indicate that elevated circulating levels of MDSCs and IL-10 may be predictive of poor functional healing outcomes and represent novel targets for immunotherapeutic intervention.


Assuntos
Biomarcadores/sangue , Regeneração Óssea/fisiologia , Fraturas não Consolidadas/imunologia , Células Supressoras Mieloides/imunologia , Animais , Quimiocinas/sangue , Quimiocinas/imunologia , Citocinas/sangue , Feminino , Fêmur/lesões , Fraturas não Consolidadas/diagnóstico por imagem , Fraturas não Consolidadas/fisiopatologia , Fraturas não Consolidadas/terapia , Imunidade/fisiologia , Interleucina-10/sangue , Interleucina-10/imunologia , Análise Multivariada , Ratos Sprague-Dawley , Microtomografia por Raio-X
20.
J Mech Behav Biomed Mater ; 116: 104380, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33588248

RESUMO

Despite the innate ability for bone to remodel and repair, its regeneration has a limit. In these cases of critically sized bone defects (CSBD), the bone deficit must be repaired using reconstructive techniques that support immediate load bearing and encourage bone bridging across the defect. High-strength porous titanium implants offer a solution for treatment of CSBD in which the scaffold can support physiological loads, provide a matrix to guide ingrowth, and carry graft materials and/or biologics. Fabrication of titanium meta-materials via additive manufacturing (AM) has unlocked the potential to modulate mechanical and biological performance to achieve a combination of properties previously unachievable. Meta-material scaffolds with topology based on triply periodic minimal surfaces (TPMS) have gained increasing interest for use in biomedical applications due to their bioinspired nature. Despite enthusiasm for TPMS-based titanium scaffolds due to their high strength to stiffness ratio, high permeability, and curvature similar to trabecular bone, there is little preclinical evidence to support their in vivo response in bone. The present study sought to evaluate the performance of gyroid-sheet titanium scaffolds produced via AM to repair a critically size femoral cortical bone defect in rats. Empty gyroid-sheet scaffolds were shown to repair segmental defects with up to 38% of torsional strength and 54% torsional stiffness of the intact femur (control) at 12-weeks. Gyroid-sheet scaffolds carrying recombinant bone morphogenic protein-2 demonstrated bridging bone growth across the length of the defect, with torsional strength and stiffness superior to that of the intact controls.


Assuntos
Fêmur , Titânio , Animais , Osso e Ossos , Porosidade , Próteses e Implantes , Ratos , Alicerces Teciduais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...