Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(2)2024 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-38279286

RESUMO

For decades, surgeons have utilized 0.9% normal saline (NS) for joint irrigation to improve visualization during arthroscopic procedures. This continues despite mounting evidence that NS exposure impairs chondrocyte metabolism and compromises articular cartilage function. We hypothesized that chondrocyte oxidative stress induced by low pH is the dominant factor driving NS toxicity, and that buffering NS to increase its pH would mitigate these effects. Effects on chondrocyte viability, reactive oxygen species (ROS) production, and overall metabolic function were assessed. Even brief exposure to NS caused cell death, ROS overproduction, and disruption of glycolysis, pentose phosphate, and tricarboxylic acid (TCA) cycle pathways. NS also stimulated ROS overproduction in synovial cells that could adversely alter the synovial function and subsequently the entire joint health. Buffering NS with 25 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) significantly increased chondrocyte viability, reduced ROS production, and returned metabolite levels to near control levels while also reducing ROS production in synovial cells. These results confirm that chondrocytes and synoviocytes are vulnerable to insult from the acidic pH of NS and demonstrate that adding a buffering agent to NS averts many of its most harmful effects.


Assuntos
Cartilagem Articular , Condrócitos , Condrócitos/metabolismo , Solução Salina , Espécies Reativas de Oxigênio/metabolismo , Estresse Oxidativo/fisiologia , Morte Celular , Cartilagem Articular/metabolismo
2.
Spine J ; 23(10): 1471-1484, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37187251

RESUMO

BACKGROUND CONTEXT: Excessive production of epidural fibrosis in the nerve root can be a pain source after laminectomy. Pharmacotherapy is a minimally invasive treatment option to attenuate epidural fibrosis by suppressing proliferation and activation of fibroblasts, inflammation, and angiogenesis, and inducing apoptosis. PURPOSE: We reviewed and tabulated pharmaceuticals with their respective signaling axes implicated in reducing epidural fibrosis. Additionally, we summarized current literature for the feasibility of novel biologics and microRNA to lessen epidural fibrosis. STUDY DESIGN/SETTING: Systematic Review. METHODS: According to the PRISMA guidelines, we systematically reviewed the literature in October 2022. The exclusion criteria included duplicates, nonrelevant articles, and insufficient detail of drug mechanism. RESULTS: We obtained a total of 2,499 articles from PubMed and Embase databases. After screening the articles, 74 articles were finally selected for the systematic review and classified based on the functions of drugs and microRNAs which included inhibition of fibroblast proliferation and activation, pro-apoptosis, anti-inflammation, and antiangiogenesis. In addition, we summarized various pathways to prevent epidural fibrosis. CONCLUSION: This study allows a comprehensive review of pharmacotherapies to prevent epidural fibrosis during laminectomy. CLINICAL SIGNIFICANCE: We expect that our review would enable researchers and clinicians to better understand the mechanism of anti-fibrosis drugs for the clinical application of epidural fibrosis therapies.


Assuntos
Laminectomia , MicroRNAs , Animais , Laminectomia/efeitos adversos , Fibrose , Apoptose , Modelos Animais , Espaço Epidural/patologia
3.
Int J Mol Sci ; 24(1)2022 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-36613910

RESUMO

Exosomes have attracted attention due to their ability to promote intercellular communication leading to enhanced cell recruitment, lineage-specific differentiation, and tissue regeneration. The object of this study was to determine the effect of exosomes on cell homing and angiogenic differentiation for pulp regeneration. Exosomes (DPSC-Exos) were isolated from rabbit dental pulp stem cells cultured under a growth (Exo-G) or angiogenic differentiation (Exo-A) condition. The characterization of exosomes was confirmed by nanoparticle tracking analysis and an antibody array. DPSC-Exos significantly promoted cell proliferation and migration when treated with 5 × 108/mL exosomes. In gene expression analysis, DPSC-Exos enhanced the expression of angiogenic markers including vascular endothelial growth factor A (VEGFA), Fms-related tyrosine kinase 1 (FLT1), and platelet and endothelial cell adhesion molecule 1 (PECAM1). Moreover, we identified key exosomal microRNAs in Exo-A for cell homing and angiogenesis. In conclusion, the exosome-based cell homing and angiogenic differentiation strategy has significant therapeutic potential for pulp regeneration.


Assuntos
Exossomos , Fator A de Crescimento do Endotélio Vascular , Animais , Coelhos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Polpa Dentária , Exossomos/metabolismo , Neovascularização Fisiológica/genética , Regeneração , Diferenciação Celular/genética , Proliferação de Células/genética
4.
Hum Gene Ther ; 33(9-10): 529-540, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34610749

RESUMO

Lubricin, a glycoprotein encoded by the proteoglycan 4 (PRG4) gene, is an essential boundary lubricant that reduces friction between articular cartilage surfaces. The loss of lubricin subsequent to joint injury plays a role in the pathogenesis of posttraumatic osteoarthritis. In this study, we describe the development and evaluation of an adeno-associated virus (AAV)-based PRG4 gene therapy intended to restore lubricin in injured joints. The green fluorescent protein (GFP) gene was inserted the PRG4 gene to facilitate tracing the distribution of the transgene product (AAV-PRG4-GFP) in vivo. Transduction efficiency of AAV-PRG4-GFP was evaluated in joint cells, and the conditioned medium containing secreted PRG4-GFP was used for shear loading/friction and viability tests. In vivo transduction of joint tissues following intra-articular injection of AAV-PRG4-GFP was confirmed in the mouse stifle joint in a surgical model of destabilization of the medial meniscus (DMM), and chondroprotective activity was tested in a rabbit anterior cruciate ligament transection (ACLT) model. In vitro studies showed that PRG4-GFP has lubricin-like cartilage-binding and antifriction properties. Significant cytoprotective effects were seen when cartilage was soaked in PRG4-GFP before cyclic shear loading (n = 3). Polymerase chain reaction and confocal microscopy confirmed the presence of PRG4-GFP DNA and protein, respectively, in a mouse DMM (n = 3 per group). In the rabbit ACLT model, AAV-PRG4-GFP gene therapy enhanced lubricin expression (p = 0.001 vs. AAV-GFP: n = 7-14) and protected the cartilage from degeneration (p = 0.014 vs. AAV-GFP: n = 9-10) when treatments were administered immediately postoperation, but efficacy was lost when treatment was delayed for 2 weeks. AAV-PRG4-GFP gene therapy protected cartilage from degeneration in a rabbit ACLT model; however, data from the ACLT model suggest that early intervention is essential for efficacy.


Assuntos
Cartilagem Articular , Osteoartrite , Animais , Cartilagem Articular/metabolismo , Dependovirus/genética , Terapia Genética , Camundongos , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/terapia , Proteoglicanas/genética , Coelhos
5.
J Pharm Sci ; 111(5): 1379-1390, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34563533

RESUMO

A mitochondrial electron transport chain member complex I inhibitor, amobarbital, can reduce oxidative damage and chondrocyte death, eventually preventing post-traumatic osteoarthritis (PTOA). Viscosupplementation using a crosslinked hyaluronic acid (HA) hydrogel is currently applied clinically for knee OA pain relief. In this work, we utilized the HA hydrogel as a drug delivery vehicle to improve the long-term efficacy of amobarbital. Here we evaluated the pharmaceutic stability of amobarbital when dispersed in a crosslinked HA hydrogel formulated in proportions intended for clinical use. We validated a high-performance liquid chromatography with an ultraviolet detector (HPLC-UV) method following International Conference for Harmonization Q2(R1) guidelines to ensure its suitability for amobarbital detection. The feasibility of this formulation's drug delivery capability was proven by measuring the release, solubility, and drug uniformity. The amobarbital/HA hydrogel showed comparable amobarbital stability in different biological fluids compared to amobarbital solution. In addition, the amobarbital/HA hydrogel imparted significantly greater drug stability when stored at 70°C for 24 hours. In conclusion, we confirmed the pharmaceutical stability of the amobarbital/HA hydrogel in various conditions and biological fluids using a validated HPLC-UV method. This data provides essential evidence in support of the use of this amobarbital/HA formulation in future clinical trials for PTOA treatment.


Assuntos
Ácido Hialurônico , Osteoartrite , Amobarbital/uso terapêutico , Cromatografia Líquida de Alta Pressão , Humanos , Ácido Hialurônico/química , Hidrogéis/uso terapêutico , Osteoartrite/tratamento farmacológico , Osteoartrite/prevenção & controle , Dor/tratamento farmacológico
6.
Spine J ; 21(6): 1021-1030, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33610806

RESUMO

BACKGROUND: Mounting evidence that oxidative stress contributes to the pathogenesis of intervertebral disc (IVD) degeneration (IDD) suggests that therapies targeting oxidative stress may slow or prevent disease progression. PURPOSE: The objective of this study was to investigate the inhibitory effects of amobarbital (Amo) on the mitochondria of nucleus pulposus (NP) cells under tert-butyl hydrogen peroxide (tBHP)-induced oxidative stress or in NP tissues under oxidative stress from tissue injury as a means of identifying therapeutic targets for IDD. STUDY DESIGN/SETTING: We tested the effects inhibiting mitochondria, a major source of oxidants, with Amo in NP cells subjected to two different forms of insult: exposure to tBHP, and physical injury induced by disc transection. N-acetylcysteine (NAC), an antioxidant known to protect NP cells, was compared to the complex I inhibitor, Amo. METHODS: NP cells were pre-treated for 2 hours with Amo, NAC, or both, and then exposed to tBHP for 1 hour. Apoptosis, necrosis, and reactive oxygen species (ROS) production were assessed using confocal microscopy and fluorescent probes (Annexin V, propidium iodide, and MitoSox Red, respectively). The activation of mitogen-activated protein kinases (MAPKs) involved in oxidative stress responses were interrogated by confocal imaging of immunofluorescence stains using phospho-specific antibodies to extracellular signal-regulated kinase (ERK), c-JUN N-terminal kinase (JNK), and p38. Mitochondrial function was assessed by imaging JC-1 staining, a probe for membrane potential. RESULTS: Amo was modestly more protective than NAC by some measures, while both agents improved mitochondrial function and lowered tBHP-induced apoptosis, necrosis, and ROS production. Activation of MAPK by tBHP was significantly suppressed by both drugs. Physically injured IVDs were treated immediately after transection with Amo or NAC for 24 hours, and then stained with dihydroethidium (DHE), a fluorescent probe for ROS production. Immunofluorescence was used to track the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a transcription factor that induces the expression of antioxidant genes. Amo and NAC significantly reduced ROS production and increased Nrf2 expression. CONCLUSION: These findings suggest that the progression of IDD may be forestalled by Amo via protection of NP cells from oxidative stress following IVD injury. CLINICAL SIGNIFICANCE: This study will define the extent to which a novel, minimally invasive procedure targeting oxidative stress in NP cells can augment surgical interventions intended to retard IVD degeneration.


Assuntos
Degeneração do Disco Intervertebral , Disco Intervertebral , Núcleo Pulposo , Preparações Farmacêuticas , Amobarbital/metabolismo , Apoptose , Humanos , Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/tratamento farmacológico , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/prevenção & controle , Estresse Oxidativo , Preparações Farmacêuticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
7.
PLoS One ; 15(12): e0244540, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33378390

RESUMO

Despite dramatic improvements in outcomes arising from the introduction of targeted therapies and immunotherapies, metastatic melanoma is a highly resistant form of cancer with 5 year survival rates of <35%. Drug resistance is frequently reported to be associated with changes in oxidative metabolism that lead to malignancy that is non-responsive to current treatments. The current report demonstrates that triphenylphosphonium(TPP)-based lipophilic cations can be utilized to induce cytotoxicity in pre-clinical models of malignant melanoma by disrupting mitochondrial metabolism. In vitro experiments demonstrated that TPP-derivatives modified with aliphatic side chains accumulated in melanoma cell mitochondria; disrupted mitochondrial metabolism; led to increases in steady-state levels of reactive oxygen species; decreased total glutathione; increased the fraction of glutathione disulfide; and caused cell killing by a thiol-dependent process that could be rescued by N-acetylcysteine. Furthermore, TPP-derivative-induced melanoma toxicity was enhanced by glutathione depletion (using buthionine sulfoximine) as well as inhibition of thioredoxin reductase (using auranofin). In addition, there was a structure-activity relationship between the aliphatic side-chain length of TPP-derivatives (5-16 carbons), where longer carbon chains increased melanoma cell metabolic disruption and cell killing. In vivo bio-distribution experiments showed that intratumoral administration of a C14-TPP-derivative (12-carbon aliphatic chain), using a slow-release thermosensitive hydrogel as a delivery vehicle, localized the drug at the melanoma tumor site. There, it was observed to persist and decrease the growth rate of melanoma tumors. These results demonstrate that TPP-derivatives selectively induce thiol-dependent metabolic oxidative stress and cell killing in malignant melanoma and support the hypothesis that a hydrogel-based TPP-derivative delivery system could represent a therapeutic drug-delivery strategy for melanoma.


Assuntos
Auranofina/administração & dosagem , Butionina Sulfoximina/administração & dosagem , Melanoma/tratamento farmacológico , Mitocôndrias/metabolismo , Compostos Organofosforados/administração & dosagem , Animais , Auranofina/farmacologia , Butionina Sulfoximina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada , Sinergismo Farmacológico , Feminino , Humanos , Hidrogéis/química , Melanoma/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Compostos Organofosforados/química , Compostos Organofosforados/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Relação Estrutura-Atividade , Temperatura , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Orthop Res ; 38(3): 629-638, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31692083

RESUMO

Joint stiffness due to fibrosis/capsule contracture is a seriously disabling complication of articular injury that surgical interventions often fail to completely resolve. Fibrosis/contracture is associated with the abnormal persistence of myofibroblasts, which over-produce and contract collagen matrices. We hypothesized that intra-articular therapy with drugs targeting myofibroblast survival (sulfasalazine), or collagen production (ß-aminopropionitrile and cis-hydroxyproline), would reduce joint stiffness in a rabbit model of fibrosis/contracture. Drugs were encapsulated in poly[lactic-co-glycolic] acid pellets and implanted in joints after fibrosis/contracture induction. Capsule α-smooth muscle actin (α-SMA) expression and intimal thickness were evaluated by immunohistochemistry and histomorphometry, respectively. Joint stiffness was quantified by flexion-extension testing. Drawer tests were employed to determine if the drugs induced cruciate ligament laxity. Joint capsule fibroblasts were tested in vitro for contractile activity and α-SMA expression. Stiffness in immobilized joints treated with blank pellets (control) was significantly higher than in non-immobilized, untreated joints (normal) (p = 0.0008), and higher than in immobilized joints treated with sulfasalazine (p = 0.0065). None of the drugs caused significant cruciate ligament laxity. Intimal thickness was significantly lower than control in the normal and sulfasalazine-treated groups (p = 0.010 and 0.025, respectively). Contractile activity in the cells from controls was significantly increased versus normal (p = 0.001). Sulfasalazine and ß-aminopropionitrile significantly inhibited this effect (p = 0.005 and 0.0006, respectively). α-SMA expression was significantly higher in control versus normal (p = 0.0021) and versus sulfasalazine (p = 0.0007). These findings support the conclusion that sulfasalazine reduced stiffness by clearing myofibroblasts from fibrotic joints. Statement of clinical significance: The results provide proof-of-concept that established joint stiffness can be resolved non-surgically. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:629-638, 2020.


Assuntos
Artropatias/tratamento farmacológico , Artropatias/patologia , Sulfassalazina/farmacologia , Aminopropionitrilo/química , Animais , Colágeno/química , Contratura/patologia , Modelos Animais de Doenças , Fibrose , Hidroxiprolina/química , Cápsula Articular/patologia , Masculino , Contração Muscular , Miofibroblastos/fisiologia , Coelhos , Estresse Mecânico
9.
Adv Healthc Mater ; 8(2): e1801236, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30556348

RESUMO

Tissue engineering potentially offers new treatments for disorders of the temporomandibular joint which frequently afflict patients. Damage or disease in this area adversely affects masticatory function and speaking, reducing patients' quality of life. Effective treatment options for patients suffering from severe temporomandibular joint disorders are in high demand because surgical options are restricted to removal of damaged tissue or complete replacement of the joint with prosthetics. Tissue engineering approaches for the temporomandibular joint are a promising alternative to the limited clinical treatment options. However, tissue engineering is still a developing field and only in its formative years for the temporomandibular joint. This review outlines the anatomical and physiological characteristics of the temporomandibular joint, clinical management of temporomandibular joint disorder, and current perspectives in the tissue engineering approach for the temporomandibular joint disorder. The tissue engineering perspectives have been categorized according to the primary structures of the temporomandibular joint: the disc, the mandibular condyle, and the glenoid fossa. In each section, contemporary approaches in cellularization, growth factor selection, and scaffold fabrication strategies are reviewed in detail along with their achievements and challenges.


Assuntos
Transtornos da Articulação Temporomandibular/cirurgia , Articulação Temporomandibular , Engenharia Tecidual/métodos , Animais , Cavidade Glenoide/anatomia & histologia , Cavidade Glenoide/citologia , Cavidade Glenoide/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Côndilo Mandibular/anatomia & histologia , Côndilo Mandibular/citologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Articulação Temporomandibular/anatomia & histologia , Articulação Temporomandibular/fisiologia , Disco da Articulação Temporomandibular/anatomia & histologia , Disco da Articulação Temporomandibular/citologia , Transtornos da Articulação Temporomandibular/diagnóstico , Transtornos da Articulação Temporomandibular/etiologia , Alicerces Teciduais
10.
J Orthop Res ; 36(9): 2439-2449, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29667227

RESUMO

Our group employed the mouse closed intra-articular fracture (IAF) model to test the hypothesis that the innate immune system plays a role in initiating synovitis and post-traumatic osteoarthritis (PTOA) in fractured joints. A transgenic strategy featuring knockout of the receptor for advanced glycation end-products (RAGE -/- ) was pursued. The 42 and 84 mJ impacts used to create fractures were in the range previously reported to cause PTOA at 60 days post-fracture. MicroCT (µCT) was used to assess fracture patterns and epiphyseal and metaphyseal bone loss at 30 and 60 days post-fracture. Cartilage degeneration, synovitis, and matrix metalloproteinase (MMP-3, -13) expression were evaluated by histologic analyses. In wild-type mice, µCT imaging showed that 84 mJ impacts led to significant bone loss at 30 days (p < 0.05), but recovered to normal at 60 days. Bone losses did not occur in RAGE-/- mice. Synovitis was significantly elevated in 84 mJ impact wild-type mice at both endpoints (30 day, p = 0.001; 60 day, p = 0.05), whereas in RAGE-/- mice synovitis was elevated only at 30 days (p = 0.02). Mankin scores were slightly elevated in both mouse strains at 30 days, but not at 60 days. Immunohistochemistry revealed significant fracture-related increases in MMP-3 and -13 expression at 30 days (p < 0.05), with no significant difference between genotypes. These findings indicated that while RAGE -/- accelerated recovery from fracture and diminished synovitis, arthritic changes were temporary and too modest to detect an effect on the pathogenesis of PTOA. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2439-2449, 2018.


Assuntos
Densidade Óssea , Receptor para Produtos Finais de Glicação Avançada/genética , Sinovite/metabolismo , Fraturas da Tíbia/patologia , Animais , Cartilagem Articular/patologia , Modelos Animais de Doenças , Fraturas Intra-Articulares , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoartrite/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Microtomografia por Raio-X
11.
Sci Transl Med ; 10(427)2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29437147

RESUMO

We tested whether inhibiting mechanically responsive articular chondrocyte mitochondria after severe traumatic injury and preventing oxidative damage represent a viable paradigm for posttraumatic osteoarthritis (PTOA) prevention. We used a porcine hock intra-articular fracture (IAF) model well suited to human-like surgical techniques and with excellent anatomic similarities to human ankles. After IAF, amobarbital or N-acetylcysteine (NAC) was injected to inhibit chondrocyte electron transport or downstream oxidative stress, respectively. Effects were confirmed via spectrophotometric enzyme assays or glutathione/glutathione disulfide assays and immunohistochemical measures of oxidative stress. Amobarbital or NAC delivered after IAF provided substantial protection against PTOA at 6 months, including maintenance of proteoglycan content, decreased histological disease scores, and normalized chondrocyte metabolic function. These data support the therapeutic potential of targeting chondrocyte metabolism after injury and suggest a strong role for mitochondria in mediating PTOA.


Assuntos
Fraturas Intra-Articulares/metabolismo , Fraturas Intra-Articulares/prevenção & controle , Mitocôndrias/metabolismo , Animais , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Feminino , Masculino , Osteoartrite/metabolismo , Osteoartrite/prevenção & controle , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Suínos
12.
Ultrasound Med Biol ; 44(4): 831-839, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29361373

RESUMO

Melanoma is one of the most aggressive types of cancer, and its incidence has increased rapidly in the past few decades. In this study, we investigated a novel treatment approach, the use of low-intensity ultrasound (2.3 W/cm2 at 1 MHz)-mediated Optison microbubble (MB) destruction (UMMD) to treat melanoma in a flank tumor model. The effect of UMMD was first evaluated in the melanoma cell line B16 F10 (B16) in vitro and then in mice inoculated with B16 cells. MB+B16 cells were exposed to US in vitro, resulting in significant cell death proportional to duty cycle (R2 = 0.74): approximately 30%, 50%, 80% and 80% cell death at 10%, 30%, 50% and 100% DC respectively. Direct implantation of tumors with MBs, followed by sonication, resulted in retarded tumor growth and improved survival (p = 0.0106). Immunohistochemical analyses confirmed the significant changes in expression of the cell proliferation marker Ki67 (p = 0.037) and a microtubule-associated protein 2 (p = 0.048) after US + MB treatment. These results suggest that UMMD could be used as a possible treatment approach in isolated melanoma and has the potential to translate to clinical trials.


Assuntos
Albuminas/farmacologia , Meios de Contraste/farmacologia , Fluorocarbonos/farmacologia , Melanoma/terapia , Microbolhas , Ultrassom/métodos , Animais , Morte Celular , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL
13.
Int J Pharm ; 539(1-2): 139-146, 2018 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-29353083

RESUMO

Melanoma is an incurable disease for which alternative treatments to chemotherapy alone are sought. Here, using a melanoma model, we investigated the antitumor potential of combining ultrasound (US) with poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with doxorubicin (DOX). The aim was to achieve synergistic tumoricidal activity through direct and indirect US-mediated damage of tumor cells combined with sustained and potentially controllable release (when combined with US) of DOX from microspheres. An in vitro release assay demonstrated an ability of US to affect the release kinetics of DOX from DOX-loaded PLGA microspheres by inducing a 12% increase in the rate of release. In vitro viability assays demonstrated that combining US with DOX-loaded PLGA microspheres resulted in synergistic tumor cell (B16-F10 melanoma cells) killing. Melanoma-bearing mice were treated intratumorally with DOX (8 µg)-loaded microspheres and subjected to US treatment at the tumor site. This treatment could significantly extend survival (mean survival (MS) = 22.1 days) compared to untreated mice (MS = 10.4 days) and most other treatments, such as blank microspheres plus US (MS = 11.5 days) and DOX (8 µg)-loaded microspheres alone (MS = 13 days). The findings that immune checkpoint blockade did not significantly extend survival of mice treated with DOX (8 µg)-loaded microspheres plus US, and that tumor-free ("cured") mice were not protected from subsequent tumor rechallenge suggests minimal involvement of the adaptive immune response in the observed antitumor activity. Nevertheless, the synergistic increase in survival of melanoma-challenged mice treated with the combination of US and DOX-loaded microspheres implicates such a treatment methodology as a promising additional tool for combatting otherwise currently incurable cancers.


Assuntos
Doxorrubicina/uso terapêutico , Melanoma/tratamento farmacológico , Melanoma/terapia , Microesferas , Terapia por Ultrassom , Animais , Linhagem Celular Tumoral , Terapia Combinada , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Liberação Controlada de Fármacos , Injeções Intralesionais , Ácido Láctico/química , Camundongos , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Análise de Sobrevida
14.
Int J Biol Macromol ; 106: 1211-1217, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28851638

RESUMO

In this study, we investigated the low intensity ultrasound (US)-controlled delivery of local cytotoxicity and drug release via induced destruction and degradation of microparticles (MPs) made of poly(lactic-co-glycolic acid) (PLGA). This study was conducted in vitro with potential application towards tumor treatment in conjunction with direct injection. MPs, either loaded with or without doxorubicin (DOX), were prepared using a double-emulsion solvent-evaporation technique. First, the MPs were exposed to US with duty cycle (DC)-modulation. The destruction and degradation of MPs were evaluated using light and scanning electron microscopy. Second, the effects of US-mediated destruction/degradation of MPs on the local cytotoxicity as well as DOX release were evaluated. US-triggered MP destruction/degradation significantly enhanced nearby cell death and DOX release. These affects occurred in proportion to the DC. Our findings indicate that controlled cytotoxicity and DOX release by US could be useful in developing the minimally invasive therapeutic applications for tumor treatment.


Assuntos
Doxorrubicina/química , Sistemas de Liberação de Medicamentos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Doxorrubicina/uso terapêutico , Liberação Controlada de Fármacos/efeitos dos fármacos , Emulsões/química , Emulsões/uso terapêutico , Humanos , Ácido Láctico/química , Ácido Láctico/uso terapêutico , Microscopia Eletrônica de Varredura , Nanopartículas/uso terapêutico , Ácido Poliglicólico/química , Ácido Poliglicólico/uso terapêutico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
15.
J Orthop Res ; 35(9): 1966-1972, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27813166

RESUMO

Serious meniscus injuries seldom heal and increase the risk for knee osteoarthritis; thus, there is a need to develop new reparative therapies. In that regard, stimulating tissue regeneration by autologous stem/progenitor cells has emerged as a promising new strategy. We showed previously that migratory chondrogenic progenitor cells (CPCs) were recruited to injured cartilage, where they showed a capability in situ tissue repair. Here, we tested the hypothesis that the meniscus contains a similar population of regenerative cells. Explant studies revealed that migrating cells were mainly confined to the red zone in normal menisci: However, these cells were capable of repopulating defects made in the white zone. In vivo, migrating cell numbers increased dramatically in damaged meniscus. Relative to non-migrating meniscus cells, migrating cells were more clonogenic, overexpressed progenitor cell markers, and included a larger side population. Gene expression profiling showed that the migrating population was more similar to CPCs than other meniscus cells. Finally, migrating cells equaled CPCs in chondrogenic potential, indicating a capacity for repair of the cartilaginous white zone of the meniscus. These findings demonstrate that, much as in articular cartilage, injuries to the meniscus mobilize an intrinsic progenitor cell population with strong reparative potential. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1966-1972, 2017.


Assuntos
Células-Tronco Adultas/fisiologia , Meniscos Tibiais/citologia , Regeneração , Animais , Bovinos , Movimento Celular , Cabras , Meniscos Tibiais/fisiologia
16.
Arthritis Rheumatol ; 67(5): 1274-85, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25623441

RESUMO

OBJECTIVE: Articular cartilage damage after joint trauma seldom heals and often leads to osteoarthritis. We previously identified a migratory chondrogenic progenitor cell (CPC) population that responds chemotactically to cell death and rapidly repopulates the injured cartilage matrix, which suggests a potential approach for articular cartilage repair. This study was undertaken to determine whether recombinant human stromal cell-derived factor 1α (rhSDF-1α), a potent CPC chemoattractant, would improve the quality of cartilage regeneration, hypothesizing that increased recruitment of CPCs by rhSDF-1α would promote the formation of cartilage matrix upon chondrogenic induction. METHODS: Full-thickness bovine chondral defects were filled with hydrogel, composed of fibrin and hyaluronic acid and containing rhSDF-1α. Cell migration was monitored, followed by chondrogenic induction. Regenerated tissue was evaluated by histology, immunohistochemistry, and scanning electron microscopy. Push-out tests and unconfined compression tests were performed to assess the strength of tissue integration and the mechanical properties of the regenerated cartilage. RESULTS: Use of rhSDF-1α dramatically improved CPC recruitment to the chondral defects at 12 days. After 6 weeks under chondrogenic conditions, cell morphology, proteoglycan density, and the ultrastructure of the repair tissue were all similar to that found in native cartilage. Compared with empty controls, neocartilage generated in rhSDF-1α-containing defects showed significantly greater interfacial strength, and acquired mechanical properties comparable to those of native cartilage. CONCLUSION: This study showed that stimulating local CPC recruitment prior to treatment with chondrogenic factors significantly improves the biochemical and mechanical properties of the cartilage tissue formed in chondral defects. This simple approach may be implemented in vivo as a one-step procedure by staging the release of chemokine and chondrogenic factors from within the hydrogel, which can be achieved using smart drug-delivery systems.


Assuntos
Cartilagem Articular/lesões , Quimiocina CXCL12/uso terapêutico , Fibrina/uso terapêutico , Regeneração Tecidual Guiada/métodos , Ácido Hialurônico/uso terapêutico , Hidrogel de Polietilenoglicol-Dimetacrilato/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Células-Tronco/citologia , Animais , Cartilagem Articular/patologia , Bovinos , Diferenciação Celular , Movimento Celular , Células Cultivadas , Condrócitos/citologia , Condrogênese , Humanos
17.
J Orthop Res ; 32(8): 981-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24797716

RESUMO

We showed previously that chondrogenic progenitor cells (CPCs) from the superficial zone of articular cartilage respond vigorously to cartilage wounding by responding chemotactically to cell debris, but the physiologic functions of CPCs remain unclear. To help bridge this knowledge gap we undertook a comparative analysis of gene expression in bovine CPCs, chondrocytes, synovial fibroblasts (synoviocytes), and cells isolated from synovial fluid (SFCs). Analysis of microarrays parsed the four cell types into two distinct groups, one composed only of chondrocytes and the other of CPCs, synoviocytes, and SFCs. The groups differed with respect to metalloendopeptidase, collagen, and cytokine gene expression. Quantitative PCR showed that, relative to chondrocytes, all other cells under-expressed cartilage matrix genes. CPCs significantly over-expressed genes encoding the chemokines interleukin 8 (IL8), and C-C motif ligand 2, while synoviocytes over-expressed the chemokine C-X-C motif Ligand 12. Sulfated glycosaminoglycan deposition in pellet cultures by CPCs was intermediate between chondrocytes and synoviocytes/SFCs. These results indicate that the CPC phenotype more closely resembles synoviocytes and SFCs than chondrocytes. CPCs show a tendency to over-express chemokines that promote immune cell chemotaxis, suggesting they mediate inflammation in response to cartilage wounding.


Assuntos
Condrócitos/metabolismo , Células-Tronco/metabolismo , Líquido Sinovial/metabolismo , Membrana Sinovial/citologia , Transcriptoma , Animais , Cartilagem/lesões , Bovinos , Quimiocina CCL2/biossíntese , Quimiocina CXCL12/biossíntese , Fibroblastos/metabolismo , Glicosaminoglicanos/biossíntese , Interleucina-8/biossíntese , Líquido Sinovial/citologia
18.
Ultrasound Med Biol ; 40(6): 1177-86, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24612644

RESUMO

Low-intensity pulsed ultrasound (LIPUS) has been studied frequently for its beneficial effects on the repair of injured articular cartilage. We hypothesized that these effects are due to stimulation of chondrogenic progenitor cell (CPC) migration toward injured areas of cartilage through focal adhesion kinase (FAK) activation. CPC chemotaxis in bluntly injured osteochondral explants was examined by confocal microscopy, and migratory activity of cultured CPCs was measured in transwell and monolayer scratch assays. FAK activation by LIPUS was analyzed in cultured CPCs by Western blot. LIPUS effects were compared with the effects of two known chemotactic factors: N-formyl-methionyl-leucyl-phenylalanine (fMLF) and high-mobility group box 1 (HMGB1) protein. LIPUS significantly enhanced CPC migration on explants and in cell culture assays. Phosphorylation of FAK at the kinase domain (Tyr 576/577) was maximized by 5 min of exposure to LIPUS at a dose of 27.5 mW/cm(2) and frequency of 3.5 MHz. Treatment with fMLF, but not HMBG1, enhanced FAK activation to a degree similar to that of LIPUS, but neither fMLF nor HMGB1 enhanced the LIPUS effect. LIPUS-induced CPC migration was blocked by suppressing FAK phosphorylation with a Src family kinase inhibitor that blocks FAK phosphorylation. Our results imply that LIPUS might be used to promote cartilage healing by inducing the migration of CPCs to injured sites, which could delay or prevent the onset of post-traumatic osteoarthritis.


Assuntos
Cartilagem Articular/metabolismo , Movimento Celular , Condrócitos/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Células-Tronco/metabolismo , Terapia por Ultrassom , Animais , Western Blotting , Bovinos , Condrócitos/citologia , Microscopia Confocal , Osteoartrite/prevenção & controle , Fosforilação , Células-Tronco/citologia , Cicatrização
19.
Tissue Eng Part A ; 20(13-14): 1807-14, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24428547

RESUMO

Depending on the damage extent and adjacent tissue condition in traumatic cartilage injury, it is possible to heal the tissue by resident cells. Unlike autologous chondrocyte implantation, short-term enzymatic treatment is an effective single-step procedure without extra cell expansion. Moreover, this method has been shown to significantly increase cellularity in lesion edges, resulting in enhanced integration and interfacial strength. We hypothesize that the locally digested extracellular matrix by treatment allows effortless cell migration from the adjacent tissue. Full-thickness cartilage discs and osteochondral explants were prepared from mature bovine stifle joints. These specimens were treated with collagenase in a culture medium. Two concentrations, 0.25 and 0.5 mg/mL, were used with various treating time of 10, 30, and 180 min. The cartilages were subsequently washed and cultured with fibrin hydrogel. The effect of enzymatic treatment on cell migration was apparent in both experiments of the cartilage disc and full-thickness cartilage defect model. In the disc culture, the treatment resulted in an approximately three to four times higher number of migrated cells than nontreated control. In short-term collagenase-treated groups, the proteoglycan (PG) loss was localized in the edge of tissue with minimal cell death. The treatment also accelerated cell migration in the full-thickness cartilage defects and some cells differentiated into chondrocytes with the deposit of PG. Gene expression results could support the characteristics of migrated cells, which had migratory ability and chondrogenic differentiation potential with overexpression of collagen type I and II, respectively. Based on these results, short-term enzymatic treatment, which can accelerate cell migration into traumatically injured cartilage, has great potential for clinical application.


Assuntos
Cartilagem Articular/citologia , Cartilagem Articular/fisiologia , Movimento Celular/efeitos dos fármacos , Clostridium histolyticum/enzimologia , Colagenases/farmacologia , Técnicas de Cultura de Órgãos , Regeneração/efeitos dos fármacos , Animais , Cartilagem Articular/efeitos dos fármacos , Bovinos , Morte Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Proteoglicanas/metabolismo , Fatores de Tempo
20.
Artigo em Inglês | MEDLINE | ID: mdl-23852876

RESUMO

We recently introduced a novel pluronic F127 and hyaluronic acid-based hydrogel (HG) designed to deliver a broad range of therapeutics. The reverse-thermal responsive HG exhibits physical properties that seem to be ideal for the local delivery of drug- and cell-based therapies to specific anatomic sites through percutaneous injection. However, questions related to the HG's safety and efficacy must first be addressed. To address these issues, we performed standard in vitro cytotoxicity and drug release tests and in vivo biocompatibility tests in a rat model. In addition, we determined whether the HG was an effective stem cell carrier in a rat cartilage defect model. We found that the HG showed viability and biocompatibility levels similar to those reported for F127 or hyaluronic acid alone. In vitro drug release studies with bupivacaine, a drug used clinically for local pain relief, revealed that after an initial burst bupivacaine was released continuously for 10 days. Stem cells loaded in the HG were retained in situ and stimulated cartilage regeneration in experimental defects. Taken as a whole, these findings support further efforts to develop the HG as a versatile system for the delivery of a wide range of therapeutic agents in humans. © 2013 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 2013.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...