Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
J Biomech Eng ; 144(5)2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34802060

RESUMO

Delayed long bone fracture healing and nonunion continue to be a significant socioeconomic burden. While mechanical stimulation is known to be an important determinant of the bone repair process, understanding how the magnitude, mode, and commencement of interfragmentary strain (IFS) affect fracture healing can guide new therapeutic strategies to prevent delayed healing or nonunion. Mouse models provide a means to investigate the molecular and cellular aspects of fracture repair, yet there is only one commercially available, clinically-relevant, locking intramedullary nail (IMN) currently available for studying long bone fractures in rodents. Having access to alternative IMNs would allow a variety of mechanical environments at the fracture site to be evaluated, and the purpose of this proof-of-concept finite element analysis study is to identify which IMN design parameters have the largest impact on IFS in a murine transverse femoral osteotomy model. Using the dimensions of the clinically relevant IMN as a guide, the nail material, distance between interlocking screws, and clearance between the nail and endosteal surface were varied between simulations. Of these parameters, changing the nail material from stainless steel (SS) to polyetheretherketone (PEEK) had the largest impact on IFS. Reducing the distance between the proximal and distal interlocking screws substantially affected IFS only when nail modulus was low. Therefore, IMNs with low modulus (e.g., PEEK) can be used alongside commercially available SS nails to investigate the effect of initial IFS or stability on fracture healing with respect to different biological conditions of repair in rodents.


Assuntos
Fraturas do Fêmur , Fixação Intramedular de Fraturas , Animais , Fenômenos Biomecânicos , Pinos Ortopédicos , Fraturas do Fêmur/cirurgia , Fêmur/cirurgia , Análise de Elementos Finitos , Fixação Intramedular de Fraturas/métodos , Consolidação da Fratura/fisiologia , Camundongos
2.
Artigo em Inglês | MEDLINE | ID: mdl-32340992

RESUMO

Staphylococcus aureus osteomyelitis is a debilitating infection of bone. Treatment of osteomyelitis is impaired by the propensity of invading bacteria to induce pathological bone remodeling that may limit antibiotic penetration to the infectious focus. The nonsteroidal anti-inflammatory drug diflunisal was previously identified as an osteoprotective adjunctive therapy for osteomyelitis, based on the ability of this compound to inhibit S. aureus quorum sensing and subsequent quorum-dependent toxin production. When delivered locally during experimental osteomyelitis, diflunisal significantly limits bone destruction without affecting bacterial burdens. However, because diflunisal's "quorum-quenching" activity could theoretically increase antibiotic recalcitrance, it is critically important to evaluate this adjunctive therapy in the context of standard-of-care antibiotics. The objective of this study is to evaluate the efficacy of vancomycin to treat osteomyelitis during local diflunisal treatment. We first determined that systemic vancomycin effectively reduces bacterial burdens in a murine model of osteomyelitis and identified a dosing regimen that decreases bacterial burdens without eradicating infection. Using this dosing scheme, we found that vancomycin activity is unaffected by the presence of diflunisal in vitro and in vivo Similarly, locally delivered diflunisal still potently inhibits osteoblast cytotoxicity in vitro and bone destruction in vivo in the presence of subtherapeutic vancomycin. However, we also found that the resorbable polyester urethane (PUR) foams used to deliver diflunisal serve as a nidus for infection. Taken together, these data demonstrate that diflunisal does not significantly impact standard-of-care antibiotic therapy for S. aureus osteomyelitis, but they also highlight potential pitfalls encountered with local drug delivery.


Assuntos
Diflunisal , Osteomielite , Infecções Estafilocócicas , Animais , Antibacterianos , Camundongos , Osteomielite/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus , Vancomicina/farmacologia
3.
J Cell Biochem ; 119(7): 5053-5059, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29600556

RESUMO

As the complexity of interactions between tumor and its microenvironment has become more evident, a critical need to engineer in vitro models that veritably recapitulate the 3D microenvironment and relevant cell populations has arisen. This need has caused many groups to move away from the traditional 2D, tissue culture plastic paradigms in favor of 3D models with materials that more closely replicate the in vivo milieu. Creating these 3D models remains a difficult endeavor for hard and soft tissues alike as the selection of materials, fabrication processes, and optimal conditions for supporting multiple cell populations makes model development a nontrivial task. Bone tissue in particular is uniquely difficult to model in part because of the limited availability of materials that can accurately capture bone rigidity and architecture, and also due to the dependence of both bone and tumor cell behavior on mechanical signaling. Additionally, the bone is a complex cellular microenvironment with multiple cell types present, including relatively immature, pluripotent cells in the bone marrow. This prospect will focus on the current 3D models in development to more accurately replicate the bone microenvironment, which will help facilitate improved understanding of bone turnover, tumor-bone interactions, and drug response. These studies have demonstrated the importance of accurately modelling the bone microenvironment in order to fully understand signaling and drug response, and the significant effects that model properties such as architecture, rigidity, and dynamic mechanical factors have on tumor and bone cell response.


Assuntos
Neoplasias Ósseas/metabolismo , Osso e Ossos/citologia , Animais , Microambiente Celular/fisiologia , Humanos , Modelos Biológicos , Engenharia Tecidual , Alicerces Teciduais , Microambiente Tumoral
4.
Clin Oral Implants Res ; 29(6): 592-602, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30240051

RESUMO

OBJECTIVE: The objective of this study was to test the hypothesis that a compression-resistant bone graft augmented with recombinant human morphogenetic protein-2 (rhBMP-2) will promote lateral ridge augmentation without the use of protective mesh in a canine model. MATERIALS & METHODS: Compression-resistant (CR) bone grafts were evaluated in a canine model of lateral ridge augmentation. Bilateral, right trapezoidal prism-shaped defects (13-14 mm long × 8-9 mm wide × 3-4 mm deep at the base) in 13 hounds (two defects per hound) were treated with one of four groups: (i) absorbable collagen sponge + 400 µg rhBMP-2/ml (ACS, clinical control) protected by titanium mesh, (ii) CR without rhBMP-2 (CR, negative control), (iii) CR + 200 µg rhBMP-2 (CR-L), or (iv) CR + 400 µg rhBMP-2 (CR-H). All animals were euthanized after 16 weeks. Ridge height and width and new bone formation were assessed by µCT, histology, and histomorphometry. The release kinetics of rhBMP-2 from CR bone grafts in vitro and in vivo in a femoral condyle defect model in rabbits was also evaluated. RESULTS: All four bone grafts promoted new bone formation (11-31.6 volume%) in the lateral ridge defects. For CR grafts, ridge height and width increased in a dose-responsive manner with increasing rhBMP-2 concentration. Ridge height and width measured for CR-H without the use of protective mesh was comparable to that measured for ACS with a protective mesh. CONCLUSIONS: At the same dose of rhBMP-2, an injectable, compression-resistant bone graft resulted in a comparable volume of new bone formation with the clinical control (ACS). These findings highlight the potential of compression-resistant bone grafts without the use of protective mesh for lateral ridge augmentation.


Assuntos
Aumento do Rebordo Alveolar/métodos , Transplante Ósseo/métodos , Cerâmica/uso terapêutico , Materiais Dentários/uso terapêutico , Polímeros/uso terapêutico , Processo Alveolar/diagnóstico por imagem , Animais , Proteína Morfogenética Óssea 2/uso terapêutico , Cães , Masculino , Coelhos , Proteínas Recombinantes , Microtomografia por Raio-X
5.
Curr Osteoporos Rep ; 15(4): 247-254, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28646444

RESUMO

PURPOSE OF REVIEW: Bone is a structurally unique microenvironment that presents many challenges for the development of 3D models for studying bone physiology and diseases, including cancer. As researchers continue to investigate the interactions within the bone microenvironment, the development of 3D models of bone has become critical. RECENT FINDINGS: 3D models have been developed that replicate some properties of bone, but have not fully reproduced the complex structural and cellular composition of the bone microenvironment. This review will discuss 3D models including polyurethane, silk, and collagen scaffolds that have been developed to study tumor-induced bone disease. In addition, we discuss 3D printing techniques used to better replicate the structure of bone. 3D models that better replicate the bone microenvironment will help researchers better understand the dynamic interactions between tumors and the bone microenvironment, ultimately leading to better models for testing therapeutics and predicting patient outcomes.


Assuntos
Neoplasias Ósseas , Osso e Ossos , Modelos Biológicos , Microambiente Tumoral , Colágeno , Humanos , Poliuretanos , Impressão Tridimensional , Seda , Engenharia Tecidual , Alicerces Teciduais
6.
Antimicrob Agents Chemother ; 60(9): 5322-30, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27324764

RESUMO

Staphylococcus aureus osteomyelitis is a common and debilitating invasive infection of bone. Treatment of osteomyelitis is confounded by widespread antimicrobial resistance and the propensity of bacteria to trigger pathological changes in bone remodeling that limit antimicrobial penetration to the infectious focus. Adjunctive therapies that limit pathogen-induced bone destruction could therefore limit morbidity and enhance traditional antimicrobial therapies. In this study, we evaluate the efficacy of the U.S. Food and Drug Administration-approved, nonsteroidal anti-inflammatory (NSAID) compound diflunisal in limiting S. aureus cytotoxicity toward skeletal cells and in preventing bone destruction during staphylococcal osteomyelitis. Diflunisal is known to inhibit S. aureus virulence factor production by the accessory gene regulator (agr) locus, and we have previously demonstrated that the Agr system plays a substantial role in pathological bone remodeling during staphylococcal osteomyelitis. Consistent with these observations, we find that diflunisal potently inhibits osteoblast cytotoxicity caused by S. aureus secreted toxins independently of effects on bacterial growth. Compared to commonly used NSAIDs, diflunisal is uniquely potent in the inhibition of skeletal cell death in vitro Moreover, local delivery of diflunisal by means of a drug-eluting, bioresorbable foam significantly limits bone destruction during S. aureus osteomyelitis in vivo Collectively, these data demonstrate that diflunisal potently inhibits skeletal cell death and bone destruction associated with S. aureus infection and may therefore be a useful adjunctive therapy for osteomyelitis.


Assuntos
Antibacterianos/farmacologia , Conservadores da Densidade Óssea/farmacologia , Preparações de Ação Retardada/farmacologia , Diflunisal/farmacologia , Reposicionamento de Medicamentos , Osteomielite/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Feminino , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteomielite/microbiologia , Osteomielite/patologia , Cultura Primária de Células , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/patogenicidade , Transativadores/antagonistas & inibidores , Transativadores/genética , Transativadores/metabolismo , Resultado do Tratamento
7.
J Biomech Eng ; 137(7)2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25902471

RESUMO

Biomaterial substrates composed of semi-aligned electrospun fibers are attractive supports for the regeneration of connective tissues because the fibers are durable under cyclic tensile loads and can guide cell adhesion, orientation, and gene expression. Previous studies on supported electrospun substrates have shown that both fiber diameter and mechanical deformation can independently influence cell morphology and gene expression. However, no studies have examined the effect of mechanical deformation and fiber diameter on unsupported meshes. Semi-aligned large (1.75 µm) and small (0.60 µm) diameter fiber meshes were prepared from degradable elastomeric poly(esterurethane urea) (PEUUR) meshes and characterized by tensile testing and scanning electron microscopy (SEM). Next, unsupported meshes were aligned between custom grips (with the stretch axis oriented parallel to axis of fiber alignment), seeded with C3H10T1/2 cells, and subjected to a static load (50 mN, adjusted daily), a cyclic load (4% strain at 0.25 Hz for 30 min, followed by a static tensile loading of 50 mN, daily), or no load. After 3 days of mechanical stimulation, confocal imaging was used to characterize cell shape, while measurements of deoxyribonucleic acid (DNA) content and messenger ribonucleic acid (mRNA) expression were used to characterize cell retention on unsupported meshes and expression of the connective tissue phenotype. Mechanical testing confirmed that these materials deform elastically to at least 10%. Cells adhered to unsupported meshes under all conditions and aligned with the direction of fiber orientation. Application of static and cyclic loads increased cell alignment. Cell density and mRNA expression of connective tissue proteins were not statistically different between experimental groups. However, on large diameter fiber meshes, static loading slightly elevated tenomodulin expression relative to the no load group, and tenascin-C and tenomodulin expression relative to the cyclic load group. These results demonstrate the feasibility of maintaining cell adhesion and alignment on semi-aligned fibrous elastomeric substrates under different mechanical conditions. The study confirms that cell morphology is sensitive to the mechanical environment and suggests that expression of select connective tissue genes may be enhanced on large diameter fiber meshes under static tensile loads.


Assuntos
Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Elasticidade , Células-Tronco Mesenquimais/efeitos dos fármacos , Poliuretanos/química , Poliuretanos/farmacologia , Animais , Contagem de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Teste de Materiais , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Estresse Mecânico , Propriedades de Superfície , Tenascina/genética , Resistência à Tração , Suporte de Carga
8.
Clin Orthop Relat Res ; 473(12): 3951-61, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26201421

RESUMO

BACKGROUND: Infectious complications of musculoskeletal trauma are an important factor contributing to patient morbidity. Biofilm-dispersive bone grafts augmented with D-amino acids (D-AAs) prevent biofilm formation in vitro and in vivo, but the effects of D-AAs on osteocompatibility and new bone formation have not been investigated. QUESTIONS/PURPOSES: We asked: (1) Do D-AAs hinder osteoblast and osteoclast differentiation in vitro? (2) Does local delivery of D-AAs from low-viscosity bone grafts inhibit new bone formation in a large-animal model? METHODS: Methicillin-sensitive Staphylococcus aureus and methicillin-resistant S aureus clinical isolates, mouse bone marrow stromal cells, and osteoclast precursor cells were treated with an equal mass (1:1:1) mixture of D-Pro:D-Met:D-Phe. The effects of the D-AA dose on biofilm inhibition (n = 4), biofilm dispersion (n = 4), and bone marrow stromal cell proliferation (n = 3) were quantitatively measured by crystal violet staining. Osteoblast differentiation was quantitatively assessed by alkaline phosphatase staining, von Kossa staining, and quantitative reverse transcription for the osteogenic factors a1Col1 and Ocn (n = 3). Osteoclast differentiation was quantitatively measured by tartrate-resistant acid phosphatase staining (n = 3). Bone grafts augmented with 0 or 200 mmol/L D-AAs were injected in ovine femoral condyle defects in four sheep. New bone formation was evaluated by µCT and histology 4 months later. An a priori power analysis indicated that a sample size of four would detect a 7.5% difference of bone volume/total volume between groups assuming a mean and SD of 30% and 5%, respectively, with a power of 80% and an alpha level of 0.05 using a two-tailed t-test between the means of two independent samples. RESULTS: Bone marrow stromal cell proliferation, osteoblast differentiation, and osteoclast differentiation were inhibited at D-AAs concentrations of 27 mmol/L or greater in a dose-responsive manner in vitro (p < 0.05). In methicillin-sensitive and methicillin-resistant S aureus clinical isolates, D-AAs inhibited biofilm formation at concentrations of 13.5 mmol/L or greater in vitro (p < 0.05). Local delivery of D-AAs from low-viscosity grafts did not inhibit new bone formation in a large-animal model pilot study (0 mmol/L D-AAs: bone volume/total volume = 26.9% ± 4.1%; 200 mmol/L D-AAs: bone volume/total volume = 28.3% ± 15.4%; mean difference with 95% CI = -1.4; p = 0.13). CONCLUSIONS: D-AAs inhibit biofilm formation, bone marrow stromal cell proliferation, osteoblast differentiation, and osteoclast differentiation in vitro in a dose-responsive manner. Local delivery of D-AAs from bone grafts did not inhibit new bone formation in vivo at clinically relevant doses. CLINICAL RELEVANCE: Local delivery of D-AAs is an effective antibiofilm strategy that does not appear to inhibit bone repair. Longitudinal studies investigating bacterial burden, bone formation, and bone remodeling in contaminated defects as a function of D-AA dose are required to further support the use of D-AAs in the clinical management of infected open fractures.


Assuntos
Aminoácidos/farmacologia , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Transplante Ósseo/métodos , Fêmur/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Animais , Antibacterianos/toxicidade , Biofilmes/crescimento & desenvolvimento , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Fêmur/diagnóstico por imagem , Fêmur/metabolismo , Fêmur/cirurgia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Metionina/farmacologia , Camundongos , Modelos Animais , Osseointegração/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Fenilalanina/farmacologia , Prolina/farmacologia , Carneiro Doméstico , Fatores de Tempo , Microtomografia por Raio-X
9.
Biomacromolecules ; 15(1): 75-83, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24266805

RESUMO

Electrospun meshes suffer from poor cell infiltration and limited thickness, which restrict their use to thin tissue applications. Herein, we demonstrate two complementary processes to overcome these limitations and achieve elastomeric composites that may be suitable for ligament repair. First, C3H10T1/2 mesenchymal stem cells were incorporated into electrospun meshes using a hybrid electrospinning/electrospraying process. Second, electrospun meshes were rolled and formed into composites with an interpenetrating polyethylene glycol (PEG) hydrogel network. Stiffer composites were formed from poly(lactic-co-glycolic acid) (PLGA) meshes, while softer and more elastic composites were formed from poly(ester-urethane urea) (PEUUR) meshes. As-spun PLGA and PEUUR rolled meshes had tensile moduli of 19.2 ± 1.9 and 0.86 ± 0.34 MPa, respectively, which changed to 11.6 ± 4.8 and 1.05 ± 0.39 MPa with the incorporation of a PEG hydrogel phase. In addition, cyclic tensile testing indicated that PEUUR-based composites deformed elastically to at least 10%. Finally, C3H10T1/2 cells incorporated into electrospun meshes survived the addition of the PEG phase and remained viable for up to 5 days. These results indicate that the fabricated cellularized composites are support cyclic mechanical conditioning, and have potential application in ligament repair.


Assuntos
Hidrogel de Polietilenoglicol-Dimetacrilato/química , Ligamentos/química , Células-Tronco Mesenquimais/química , Engenharia Tecidual/métodos , Animais , Linhagem Celular , Hidrogel de Polietilenoglicol-Dimetacrilato/metabolismo , Ácido Láctico/química , Ácido Láctico/metabolismo , Ligamentos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Ácido Poliglicólico/química , Ácido Poliglicólico/metabolismo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Estresse Mecânico
10.
Curr Osteoporos Rep ; 12(1): 48-54, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24458428

RESUMO

Healing fractures resulting from osteoporosis or cancer remains a significant clinical challenge. In these populations, healing is often impaired not only due to age and disease, but also by other therapeutic interventions such as radiation, steroids, and chemotherapy. Despite substantial improvements in the treatment of osteoporosis over the last few decades, osteoporotic fractures are still a major clinical challenge in the elderly population due to impaired healing. Similar fractures with impaired healing are also prevalent in cancer patients, especially those with tumor growing in bone. Treatment options for cancer patients are further complicated by the fact that bone anabolic therapies are contraindicated in patients with tumors. Therefore, many patients undergo surgery to repair the fracture, and bone grafts are often used to stabilize orthopedic implants and provide a scaffold for ingrowth of new bone. Both synthetic and naturally occurring biomaterials have been investigated as bone grafts for repair of osteoporotic fractures, including calcium phosphate bone cements, resorbable polymers, and allograft or autograft bone. In order to re-establish normal bone repair, bone grafts have been augmented with anabolic agents, such as mesenchymal stem cells or recombinant human bone morphogenetic protein-2. These developing approaches to bone grafting are anticipated to improve the clinical management of osteoporotic and cancer-induced fractures.


Assuntos
Proteína Morfogenética Óssea 2/uso terapêutico , Neoplasias Ósseas/secundário , Transplante Ósseo/métodos , Fixação de Fratura/métodos , Fraturas Espontâneas/terapia , Osteoporose/terapia , Fraturas por Osteoporose/terapia , Hormônio Paratireóideo/uso terapêutico , Alicerces Teciduais , Fator de Crescimento Transformador beta/uso terapêutico , Materiais Biocompatíveis/uso terapêutico , Neoplasias Ósseas/complicações , Fraturas Espontâneas/etiologia , Humanos , Dispositivos de Fixação Ortopédica , Proteínas Recombinantes/uso terapêutico
11.
J Mater Res ; 29(20): 2398-2407, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25798027

RESUMO

Injectable bone grafts with strength exceeding that of trabecular bone could improve the management of a number of orthopaedic conditions. Ceramic/polymer composites have been investigated as weight-bearing bone grafts, but they are typically weaker than trabecular bone due to poor interfacial bonding. We hypothesized that entrapment of surface-initiated poly(ε-caprolactone) (PCL) chains on 45S5 bioactive glass (BG) particles within an in situ-formed polymer network would enhance the mechanical properties of reactive BG/polymer composites. When the surface-initiated PCL molecular weight exceeded the molecular weight between crosslinks of the network, the compressive strength of the composites increased 6- to 10-fold. The torsional strength of the composites exceeded that of human trabecular bone by a factor of two. When injected into femoral condyle defects in rats, the composites supported new bone formation at 8 weeks. The initial bone-like strength of BG/polymer composites and their ability to remodel in vivo highlight their potential for development as injectable grafts for repair of weight-bearing bone defects.

12.
Cancer Res Commun ; 3(2): 223-234, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36968140

RESUMO

When breast cancer metastasizes to bone, treatment options are limited. Failure to treat bone metastases is thought to be due to therapy-resistant features of the bone marrow microenvironment. Using a murine model of bone metastatic mammary carcinoma, we demonstrate that systemic delivery of polymer nanoparticles loaded with cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) inhibited tumor growth and bone destruction after 7 days of treatment. Each dose of STING-activating nanoparticles trafficked to the bone marrow compartment and was retained within the tumor microenvironment for over 24 hours, enhancing antitumor immunity through proinflammatory cytokine production and early T-cell activation. While acquired resistance mechanisms, including increased levels of immunosuppressive cytokines and the infiltration of regulatory T cells, ultimately limited antitumor efficacy after 2 weeks of treatment, bone protective effects remained. Overall, these studies demonstrate that STING pathway activation, here enabled using a nanomedicine approach to enhance CDN delivery to bone metastatic sites, can reprogram the immune contexture of the bone marrow to an antitumor phenotype that inhibits bone colonization of metastatic breast cancer cells and protects from tumor-mediated bone destruction. Significance: Bone metastases are difficult to treat due to the inaccessibility of the bone marrow compartment and the immunosuppressive microenvironment that protects resident stem cells. Packaging a STING agonist into a nanoparticle that enables systemic administration and drug accumulation at tumor sites overcomes both barriers to stymie metastatic breast cancer growth.


Assuntos
Nanopartículas , Neoplasias , Camundongos , Animais , Medula Óssea , Citocinas , Fenótipo , Microambiente Tumoral
13.
Curr Protoc ; 2(1): e333, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34985830

RESUMO

Advanced solid tumors often metastasize to bone. Once established in bone, these tumors can induce bone destruction resulting in decreased quality of life and increased mortality. Neither 2D in vitro models nor 3D animal models sufficiently recapitulate the human bone-tumor microenvironment needed to fully understand the complexities of bone metastasis, highlighting the need for new models. A 3D in vitro humanized model of tumor-induced bone disease was developed by dynamically culturing human osteoblast, osteoclast, and metastatic cancer cells together within tissue-engineered bone constructs. Cell-mediated resorption can be observed by micro-computed tomography and can be quantified by change in mass. Taken together, these data can be used to investigate whether the metastatic cancer cells included in the model have the potential to drive osteoclastogenesis and cell-mediated resorption in vitro. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Fabricating bone-like scaffolds Basic Protocol 2: Preparing cells for the humanized model of TIBD Basic Protocol 3: Crafting a 3D in vitro humanized model of TIBD.


Assuntos
Neoplasias Ósseas , Qualidade de Vida , Animais , Reatores Biológicos , Humanos , Perfusão , Microambiente Tumoral , Microtomografia por Raio-X
14.
Sci Transl Med ; 14(641): eabm6586, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35442705

RESUMO

Porous, resorbable biomaterials can serve as temporary scaffolds that support cell infiltration, tissue formation, and remodeling of nonhealing skin wounds. Synthetic biomaterials are less expensive to manufacture than biologic dressings and can achieve a broader range of physiochemical properties, but opportunities remain to tailor these materials for ideal host immune and regenerative responses. Polyesters are a well-established class of synthetic biomaterials; however, acidic degradation products released by their hydrolysis can cause poorly controlled autocatalytic degradation. Here, we systemically explored reactive oxygen species (ROS)-degradable polythioketal (PTK) urethane (UR) foams with varied hydrophilicity for skin wound healing. The most hydrophilic PTK-UR variant, with seven ethylene glycol (EG7) repeats flanking each side of a thioketal bond, exhibited the highest ROS reactivity and promoted optimal tissue infiltration, extracellular matrix (ECM) deposition, and reepithelialization in porcine skin wounds. EG7 induced lower foreign body response, greater recruitment of regenerative immune cell populations, and resolution of type 1 inflammation compared to more hydrophobic PTK-UR scaffolds. Porcine wounds treated with EG7 PTK-UR foams had greater ECM production, vascularization, and resolution of proinflammatory immune cells compared to polyester UR foam-based NovoSorb Biodegradable Temporizing Matrix (BTM)-treated wounds and greater early vascular perfusion and similar wound resurfacing relative to clinical gold standard Integra Bilayer Wound Matrix (BWM). In a porcine ischemic flap excisional wound model, EG7 PTK-UR treatment led to higher wound healing scores driven by lower inflammation and higher reepithelialization compared to NovoSorb BTM. PTK-UR foams warrant further investigation as synthetic biomaterials for wound healing applications.


Assuntos
Materiais Biocompatíveis , Cicatrização , Animais , Bandagens , Materiais Biocompatíveis/farmacologia , Inflamação , Poliésteres , Espécies Reativas de Oxigênio , Pele , Suínos
15.
Biophys J ; 100(3): 573-582, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21281571

RESUMO

Recent studies have suggested that extracellular matrix rigidity regulates cancer invasiveness, including the formation of cellular invadopodial protrusions; however, the relevant mechanical range is unclear. Here, we used a combined analysis of tissue-derived model basement membrane (BM) and stromal matrices and synthetic materials to understand how substrate rigidity regulates invadopodia. Urinary bladder matrix-BM (UBM-BM) was found to be a rigid material with elastic moduli of 3-8 MPa, as measured by atomic force microscopy and low-strain tensile testing. Stromal elastic moduli were ∼6-fold lower, indicating a more compliant material. Using synthetic substrates that span kPa-GPa moduli, we found a peak of invadopodia-associated extracellular matrix degradation centered around 30 kPa, which also corresponded to a peak in invadopodia/cell. Surprisingly, we observed another peak in invadopodia numbers at 2 GPa as well as gene expression changes that indicate cellular sensing of very high moduli. Based on the measured elastic moduli of model stroma and BM, we expected to find more invadopodia formation on the stroma, and this was verified on the stromal versus BM side of UBM-BM. These data suggest that cells can sense a wide range of rigidities, up into the GPa range. Furthermore, there is an optimal rigidity range for invadopodia activity that may be limited by BM rigidity.


Assuntos
Extensões da Superfície Celular/metabolismo , Matriz Extracelular/metabolismo , Resinas Acrílicas/farmacologia , Animais , Membrana Basal/efeitos dos fármacos , Membrana Basal/metabolismo , Fenômenos Biomecânicos/efeitos dos fármacos , Extensões da Superfície Celular/efeitos dos fármacos , Módulo de Elasticidade/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Microscopia de Força Atômica , Modelos Biológicos , Poliuretanos/farmacologia , Pressão , Sus scrofa , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/metabolismo
16.
Curr Biol ; 18(17): 1295-1299, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18718759

RESUMO

Invadopodia are actin-rich subcellular protrusions with associated proteases used by cancer cells to degrade extracellular matrix (ECM) [1]. Molecular components of invadopodia include branched actin-assembly proteins, membrane trafficking proteins, signaling proteins, and transmembrane proteinases [1]. Similar structures exist in nontransformed cells, such as osteoclasts and dendritic cells, but are generally called podosomes and are thought to be more involved in cell-matrix adhesion than invadopodia [2-4]. Despite intimate contact with their ECM substrates, it is unknown whether physical or chemical ECM signals regulate invadopodia function. Here, we report that ECM rigidity directly increases both the number and activity of invadopodia. Transduction of ECM-rigidity signals depends on the cellular contractile apparatus [5-7], given that inhibition of nonmuscle myosin II, myosin light chain kinase, and Rho kinase all abrogate invadopodia-associated ECM degradation. Whereas myosin IIA, IIB, and phosphorylated myosin light chain do not localize to invadopodia puncta, active phosphorylated forms of the mechanosensing proteins p130Cas (Cas) and focal adhesion kinase (FAK) are present in actively degrading invadopodia, and the levels of phospho-Cas and phospho-FAK in invadopodia are sensitive to myosin inhibitors. Overexpression of Cas or FAK further enhances invadopodia activity in cells plated on rigid polyacrylamide substrates. Thus, in invasive cells, ECM-rigidity signals lead to increased matrix-degrading activity at invadopodia, via a myosin II-FAK/Cas pathway. These data suggest a potential mechanism, via invadopodia, for the reported correlation of tissue density with cancer aggressiveness.


Assuntos
Extensões da Superfície Celular/fisiologia , Matriz Extracelular/fisiologia , Citoesqueleto de Actina/metabolismo , Azepinas/farmacologia , Linhagem Celular Tumoral , Extensões da Superfície Celular/ultraestrutura , Proteína Substrato Associada a Crk/análise , Proteína Substrato Associada a Crk/fisiologia , Inibidores Enzimáticos/farmacologia , Matriz Extracelular/ultraestrutura , Quinase 1 de Adesão Focal/análise , Quinase 1 de Adesão Focal/fisiologia , Gelatina/química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Integrinas/metabolismo , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Naftalenos/farmacologia , Transdução de Sinais
17.
Calcif Tissue Int ; 89(2): 111-22, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21597909

RESUMO

The full range of fracture risk determinants arise from each hierarchical level comprising the organization of bone. Raman spectroscopy is one tool capable of characterizing the collagen and mineral phases at a near submicron-length scale, but the ability of Raman spectra to distinguish compositional differences of bone is not well defined. Therefore, we analyzed multiple Raman peak intensities and peak ratios to characterize their ability to distinguish between the typically less mineralized osteonal tissue and the more mineralized interstitial tissue in intracortical human bone. To further assess origins of variance, we collected Raman spectra from embedded specimens and for two orientations of cut. Per specimen, Raman peak intensities or ratios were averaged among multiple sites within five osteons and five neighboring interstitial tissue. The peak ratios of ν(1) phosphate (PO(4)) to proline or amide III detected the highest increases of 15.4 or 12.5%, respectively, in composition from osteonal to interstitial tissue. The coefficient of variance was less than 5% for each as opposed to a value of ~8% for the traditional ν(1)PO(4)/amide I, a peak ratio that varied the most between transverse and longitudinal cuts for each tissue type. Although embedding affected Raman peaks, it did not obscure differences in most peak ratios related to mineralization between the two tissue types. In studies with limited sample size but sufficient number of Raman spectra per specimen for spatial averaging, ν(1)PO(4)/amide III or ν(1)PO(4)/proline is the Raman property that is most likely to detect a compositional difference between experimental groups.


Assuntos
Densidade Óssea , Osso e Ossos/química , Análise Espectral Raman/métodos , Idoso de 80 Anos ou mais , Animais , Densidade Óssea/fisiologia , Osso e Ossos/ultraestrutura , Feminino , Humanos , Individualidade , Masculino , Camundongos , Microdissecção/métodos , Pessoa de Meia-Idade , Especificidade de Órgãos/fisiologia , Ratos , Inclusão do Tecido/métodos , Preservação de Tecido/métodos , Pesos e Medidas
18.
Curr Osteoporos Rep ; 9(2): 89-95, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21424744

RESUMO

Tumors such as breast, lung, and prostate frequently metastasize to bone, where they can cause intractable pain and increase the risk of fracture in patients. When tumor cells metastasize to bone, they interact with the microenvironment to promote bone destruction primarily through the secretion of osteolytic factors by the tumor cells and the subsequent release of growth factors from the bone. Our recent data suggest that the differential rigidity of the mineralized bone microenvironment relative to that of soft tissue regulates the expression of osteolytic factors by the tumor cells. The concept that matrix rigidity regulates tumor growth is well established in solid breast tumors, where increased rigidity stimulates tumor cell invasion and metastasis. Our studies have indicated that a transforming growth factor-ß (TGF-ß) and Rho-associated kinase (ROCK)-dependent mechanism is involved in the response of metastatic tumor cells to the rigid mineralized bone matrix. In this review, we will discuss the interactions between ROCK and TGF-ß signaling, as well as potential new therapies that target these pathways.


Assuntos
Neoplasias Ósseas , Fator de Crescimento Transformador beta/metabolismo , Quinases Associadas a rho/metabolismo , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Conservadores da Densidade Óssea/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Neoplasias Ósseas/fisiopatologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Denosumab , Difosfonatos/farmacologia , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Ligante RANK/uso terapêutico , Proteína Gli2 com Dedos de Zinco
19.
Materials (Basel) ; 14(14)2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-34300888

RESUMO

Autograft (AG) is the gold standard for bone grafts, but limited quantities and patient morbidity are associated with its use. AG extenders have been proposed to minimize the volume of AG while maintaining the osteoinductive properties of the implant. In this study, poly(ester urethane) (PEUR) and poly(thioketal urethane) (PTKUR) AG extenders were implanted in a 20-mm rabbit radius defect model to evaluate new bone formation and graft remodeling. Outcomes including µCT and histomorphometry were measured at 12 weeks and compared to an AG (no polymer) control. AG control examples exhibited new bone formation, but inconsistent healing was observed. The implanted AG control was resorbed by 12 weeks, while AG extenders maintained implanted AG throughout the study. Bone growth from the defect interfaces was observed in both AG extenders, but residual polymer inhibited cellular infiltration and subsequent bone formation within the center of the implant. PEUR-AG extenders degraded more rapidly than PTKUR-AG extenders. These observations demonstrated that AG extenders supported new bone formation and that polymer composition did not have an effect on overall bone formation. Furthermore, the results indicated that early cellular infiltration is necessary for harnessing the osteoinductive capabilities of AG.

20.
J Orthop Res ; 39(2): 426-437, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33300149

RESUMO

Osteomyelitis is a debilitating infection of bone that results in substantial morbidity. Staphylococcus aureus is the most commonly isolated pathogen causing bone infections and features an arsenal of virulence factors that contribute to bone destruction and counteract immune responses. We previously demonstrated that diflunisal, a nonsteroidal anti-inflammatory drug, decreases S. aureus-induced bone destruction during osteomyelitis when delivered locally from a resorbable drug delivery depot. However, local diflunisal therapy was complicated by bacterial colonization of the depot's surface, highlighting a common pitfall of devices for local drug delivery to infected tissue. It is, therefore, critical to develop an alternative drug delivery method for diflunisal to successfully repurpose this drug as an antivirulence therapy for osteomyelitis. We hypothesized that a nanoparticle-based parenteral delivery strategy would provide a method for delivering diflunisal to infected tissue while circumventing the complications associated with local delivery. In this study, we demonstrate that poly(propylene sulfide) (PPS) nanoparticles accumulate at the infectious focus in a murine model of staphylococcal osteomyelitis and are capable of efficaciously delivering diflunisal to infected bone. Moreover, diflunisal-loaded PPS nanoparticles effectively decrease S. aureus-mediated bone destruction, establishing the feasibility of systemic delivery of an antivirulence compound to mitigate bone pathology during osteomyelitis.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Osso e Ossos/efeitos dos fármacos , Diflunisal/administração & dosagem , Osteomielite/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Animais , Linhagem Celular , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Feminino , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Camundongos , Nanopartículas/química , Polímeros , Staphylococcus aureus , Sulfetos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA