Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
World Neurosurg ; 183: e440-e446, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38154684

RESUMO

OBJECTIVE: Lumbar spinal fusion is a common surgical procedure that can be done with a variety of different instrumentation and techniques. Despite numerous research studies investigating subsidence risk factors, the impact of cage placement on subsidence is not fully elucidated. This study aims to determine whether placement of an expandable transforaminal lumbar interbody fusion cage at the center end plate or at the anterior apophyseal ring affects cage subsidence. METHODS: A transforaminal lumbar interbody fusion cage was placed centrally or peripherally between 2 synthetic vertebral models of L3 and L4. A compression plate attached to a 10 KN load cell was used to uniaxially compress the assembly. The ultimate force required for the assembly to fail and subsidence stiffness were analyzed. Computed tomography scans of each L3 and L4 were obtained, and maximum end plate subsidence was measured in the frontal plane. RESULTS: Anterior apophyseal cage placement resulted in higher stiffness of the vertebrae-cage assembly (Ks, 962.89 N/mm) and a higher subsidence stiffness (Kb,987.21 N/mm) compared with central placement (P < 0.05). Ultimate compressive load of the vertebrae-cage assembly did not increase. Moreover, the maximum subsidence depth did not significantly vary between placements. CONCLUSIONS: The subsidence stiffness increased with anterior apophyseal cage placement. Periphery end plate cortical bone architecture may play a role in resisting the impact of cage subsidence. To fully understand the effect of cage placement on cage subsidence, future studies should investigate its implications on native and diseased spine.


Assuntos
Vértebras Lombares , Fusão Vertebral , Humanos , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/cirurgia , Fenômenos Biomecânicos , Placas Ósseas , Fusão Vertebral/métodos , Região Lombossacral
2.
Hand (N Y) ; 18(3): 436-445, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-34340572

RESUMO

BACKGROUND: As hand surgeons, tendon injuries and lacerations are a particularly difficult problem to treat, as poor healing potential and adhesions hamper optimal recovery. Adipose-derived stem cells (ADSCs) have been shown to aid in rat Achilles tendon healing after a puncture defect, and this model can be used to study tendon healing in the upper extremity. We hypothesized that ADSCs cultured with growth differentiation factor 5 (GDF5) and platelet-derived growth factor (PDGF) would improve tendon healing after a transection injury. METHODS: Rat Achilles tendons were transected and then left either unrepaired or repaired. Both groups were treated with a hydrogel alone, a hydrogel with ADSCs, or a hydrogel with ADSCs that were cultured with GDF5 and PDGF prior to implantation. Tissue harvested from the tendons was evaluated for gene expression of several genes known to play an important role in successful tendon healing. Histological examination of the tendon healing was also performed. RESULTS: In both repaired and unrepaired tendons, those treated with ADSCs cultured with GDF5/PDGF prior to implantation showed the best tendon fiber organization, the smallest gaps, and the most organized blood vessels. Treatment with GDF5/PDGF increased expression of the protenogenesis gene SOX9, promoted cell-to-cell connections, improved cellular proliferation, and enhanced tissue remodeling. CONCLUSIONS: Adipose-derived stem cells cultured with GDF5/PDGF prior to implantation can promote tendon repair by improving cellular proliferation, tenogenesis, and vascular infiltration. This effect results in a greater degree of organized tendon healing.


Assuntos
Tendão do Calcâneo , Fator de Crescimento Derivado de Plaquetas , Ratos , Animais , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator 5 de Diferenciação de Crescimento/metabolismo , Hidrogéis/metabolismo , Células-Tronco
3.
J Orthop Surg Res ; 15(1): 94, 2020 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-32138774

RESUMO

BACKGROUND: 5-Aminolevulinic acid (5-ALA), a fluorescent contrast agent, has been used for tumor paint and photodynamic therapy (PDT) for various tumors, but its use with soft tissue sarcomas is not well documented. Myxofibrosarcoma, a subtype of soft tissue sarcoma with a high local recurrence rate, may benefit from similar types of treatment. The purpose of this study was to analyze the effects of 5-ALA tumor paint and PDT on a myxofibrosarcoma cell line. METHODS: Tumor paint was assessed by exposing micromass pellets of human adipose-derived stromal (ADS) cells or myxofibrosarcoma (MUG-Myx1) cells to 5-ALA. Cell pellets were then visualized using a microscope at established excitation and emission wavelengths. Corrected total cell fluorescence was calculated per accepted protocols. Photodynamic therapy was similarly assessed by exposing ADS and MUG-Myx1 cells to 5-ALA, with subsequent analysis via flow cytometry and real-time confocal microscopy. RESULTS: The use of 5-ALA tumor paint led to a selective fluorescence in MUG-Myx1 cells. Findings were confirmed by flow cytometry. Interestingly, flow cytometry results showed progressive selective cell death with increasing 5-ALA exposure as a result of the PDT effect. PDT was further confirmed using confocal microscopy, which revealed progressive cellular bubble formation consistent with advancing stages of cell death-a finding that was not seen in control ADS cells. CONCLUSIONS: 5-ALA tumor paint and PDT were successfully used on a human myxofibrosarcoma cell line (MUG-Myx1). Results from this study showed both selective fluorescent tagging and selective cytotoxicity of 5-ALA toward malignant myxofibrosarcoma cells, while sparing benign adipose control cells. This finding was further confirmed in a dramatic time-lapse video, visually confirming active, targeted cell death. 5-ALA's two-pronged application of selective tumor identification and cytotoxicity may transform surgical and medical approaches for treating soft tissue sarcomas.


Assuntos
Ácido Aminolevulínico/toxicidade , Meios de Contraste/toxicidade , Fibroma/terapia , Fibrossarcoma/terapia , Fotoquimioterapia/métodos , Ácido Aminolevulínico/análise , Ácido Aminolevulínico/uso terapêutico , Linhagem Celular Tumoral , Meios de Contraste/análise , Meios de Contraste/uso terapêutico , Fibroma/diagnóstico , Fibrossarcoma/diagnóstico , Humanos , Microscopia Confocal/métodos
4.
Front Cell Dev Biol ; 8: 595932, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33634095

RESUMO

BACKGROUND: Microfracture is one of the most widely used techniques for the repair of articular cartilage. However, microfracture often results in filling of the chondral defect with fibrocartilage, which exhibits poor durability and sub-optimal mechanical properties. Stromal cell-derived factor-1 (SDF-1) is a potent chemoattractant for mesenchymal stem cells (MSCs) and is expressed at high levels in bone marrow adjacent to developing cartilage during endochondral bone formation. Integrating SDF-1 into an implantable collagen scaffold may provide a chondro-conductive and chondro-inductive milieu via chemotaxis of MSCs and promotion of chondrogenic differentiation, facilitating more robust hyaline cartilage formation following microfracture. OBJECTIVE: This work aimed to confirm the chemoattractive properties of SDF-1 in vitro and develop a one-step method for incorporating SDF-1 in vivo to enhance cartilage repair using a rat osteochondral defect model. METHODS: Bone marrow-derived MSCs (BMSCs) were harvested from the femurs of Sprague-Dawley rats and cultured in low-glucose Dulbecco's modified Eagle's medium containing 10% fetal bovine serum, with the medium changed every 3 days. Passage 1 MSCs were analyzed by flow cytometry with an S3 Cell Sorter (Bio-Rad). In vitro cell migration assays were performed on MSCs by labeling cells with carboxyfluorescein diacetate, succinimidyl ester (CFDA-SE; Bio-Rad). For the microfracture model, a 1.6-mm-diameter osteochondral defect was created in the femoral trochleae of 20 Sprague-Dawley rats bilaterally until bone marrow spillage was seen under saline irrigation. One knee was chosen at random to receive implantation of the scaffold, and the contralateral knee was left unfilled as an empty control. Type I collagen scaffolds (Kensey Nash) were coated with either gelatin only or gelatin and SDF-1 using a dip coating process. The rats received implantation of either a gelatin-only scaffold (N = 10) or gelatin-and-SDF-1 scaffold (N = 10) at the site of the microfracture. Femurs were collected for histological analyses at 4- and 8-week time points post-operatively, and sections were stained with Safranin O/Fast Green. The samples were graded blindly by two observers using the Modified O'Driscoll score, a validated scoring system for chondral repair. A minimum of 10 separate grading scores were made per sample and averaged. Quantitative comparisons of cell migration in vitro were performed with one-way ANOVA. Cartilage repair in vivo was also compared among groups with one-way ANOVA, and the results were presented as mean ± standard deviation, with P-values < 0.05 considered as statistically significant. RESULTS: MSC migration showed a dose-response relationship with SDF-1, with an optimal dosage for chemotaxis between 10 and 100 ng/ml. After scaffold implantation, the SDF-1-treated group demonstrated complete filling of the cartilage defect with mature cartilage tissue, exhibiting strong proteoglycan content, smooth borders, and good incorporation into marginal cartilage. Modified O'Driscoll scores after 8 weeks showed a significant improvement of cartilage repair in the SDF-1 group relative to the empty control group (P < 0.01), with a trend toward improvement when compared with the gelatin-only-scaffold group (P < 0.1). No significant differences in scores were found between the empty defect group and gelatin-only group. CONCLUSION: In this study, we demonstrated a simple method for improving the quality of cartilage defect repair in a rat model of microfracture. We confirmed the chemotactic properties of SDF-1 on rat MSCs and found an optimized dosage range for chemotaxis between 10 and 100 ng/ml. Furthermore, we demonstrated a strategy to incorporate SDF-1 into gelatin-collagen I scaffolds in vivo at the site of an osteochondral defect. SDF-1-treated defects displayed robust hyaline cartilage resurfacing of the defect with minimal fibrous tissue, in contrast to the empty control group. The results of the in vitro and in vivo studies together suggest that SDF-1-mediated signaling may significantly improve the quality of cartilage regeneration in an osteochondral defect.

5.
J Tissue Eng ; 9: 2041731418811183, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30542597

RESUMO

The purpose of this study was to characterize rat adipose-derived stem cells, induce adipose-derived stem cell tenogenesis, and analyze adipose-derived stem cell effects on tendon repair in vivo. Adipose-derived stem cells demonstrated an immunomodulatory, pro-angiogenic, and pro-proliferatory profile in vitro. Tenogenesis was induced for 1, 7, 14, and 21 days with 24 combinations of growth differentiation factor-5, 6, and 7 and platelet-derived growth factor-BB. Adipose-derived stem cells expression of scleraxis and collagen type I increased the most after 14 days of induction with growth differentiation factor-6 and platelet-derived growth factor-BB. Achilles excision defects injected with hydrogel alone (Gp2), with undifferentiated (Gp3) adipose-derived stem cells, or tenogenically differentiated (Gp4) adipose-derived stem cells exhibited improved tissue repair compared with untreated tendons (Gp1). Addition of adipose-derived stem cells improved tissue cytoarchitecture and increased expression of collagen type I and III, scleraxis, and tenomodulin. Adipose-derived stem cells significantly improved biomechanical properties (ultimate load and elastic toughness) over time more than hydrogel alone, while tenogenically differentiated adipose-derived stem cells improved the mean histological score and collagen fiber dispersion range closest to normal tendon. In addition, tendon sections treated with GFP-adipose-derived stem cells exhibited green fluorescence and positive GFP immunostaining on microscopy confirming the in vivo survival of adipose-derived stem cells that were injected into tendon defects to support the effects of adipose-derived stem cells on tissue up to 4.5 weeks post injury.

6.
Shock ; 50(3): 346-350, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29065066

RESUMO

Severe burn causes systemic inflammation and hypercatabolism, resulting in damage to multiple organs distant to the burn site, including the musculoskeletal system. Bone mass and muscle loss have been reported. However, tendon that connects bone and muscle has not been studied in comparable detail. Here we aimed to characterize the molecular and functional changes in Achilles tendon triggered by severe burn. Forty male Sprague-Dawley rats received 40% total body surface area scald burn. Achilles tendons were collected up to 14 days postburn. Sham-treated animals served as a control group. We analyzed tendons for changes in expression of IL-6, IL-1ß, TNF, MMP9, MMP13, TGFß1, Collagens I and III, and for morphological and biomechanical changes. Gene expression of IL-6 and IL-1ß as well as MMP9 and MMP13 increased in rat tendon 3 days after burn. Col3a1 increased at day 3 and col1a1 at day 7. At day 14, TGFß1 increased, whereas the protein ratio for collagens I/III decreased, indicating tendon remodeling. Histological analysis with H&E and Picrosirius red staining further revealed a decrease in organized collagen fibers 14 days after burn. Biomechanical analysis showed a decrease in stiffness and ultimate force of tendons in burn rats.We conclude that tendinopathy was observed in Achilles tendon 14 days after severe burn, via the induction of inflammation and remodeling. The present study provides a model of tendinopathy that may be used for the development of therapeutic approaches after burn.


Assuntos
Tendão do Calcâneo/metabolismo , Queimaduras/metabolismo , Tendão do Calcâneo/patologia , Animais , Queimaduras/patologia , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Inflamação/patologia , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Ratos , Ratos Sprague-Dawley , Índices de Gravidade do Trauma
7.
Cartilage ; 9(4): 438-449, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-28399641

RESUMO

Objective To establish whether a novel biomaterial scaffold with tunable degradation profile will aid in cartilage repair of chondral defects versus microfracture alone in vitro and in a rat model in vivo. Design In vitro-Short- and long-term degradation scaffolds were seeded with culture expanded articular chondrocytes or bone marrow mesenchymal stem cells. Cell growth and differentiation were evaluated with cell morphological studies and gene expression studies. In vivo-A microfracture rat model was used in this study to evaluate the repair of cartilage and subchondral bone with the contralateral knee serving as the empty control. The treatment groups include (1) empty osteochondral defect, (2) polycaprolactone copolymer-based polyester polyurethane-urea (PSPU-U) caffold short-term degradative profile, and (3) PSPU-U scaffold long-term degradative profile. After placement of the scaffold, the rats were then allowed unrestricted activity as tolerated, and histological analyses were performed at 4, 8, and 16 weeks. The cartilage defect was measured and compared with the contralateral control side. Results In vitro-Long-term scaffolds showed statistically significant higher levels of aggrecan and type II collagen expression compared with short-term scaffolds. In vivo-Within 16 weeks postimplantation, there was new subchondral bone formation in both scaffolds. Short-term scaffolds had a statistically significant increase in defect filling and better qualitative histologic fill compared to control. Conclusions The PSPU short-term degradation scaffold may aid in cartilage repair by ultimately incorporating the scaffold into the microfracture procedure.


Assuntos
Doenças das Cartilagens/cirurgia , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Cartilagem Articular/citologia , Cartilagem Articular/cirurgia , Fêmur , Células-Tronco Mesenquimais/metabolismo , Ratos , Tíbia
8.
Polym Adv Technol ; 27(8): 1050-1055, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27587941

RESUMO

The hemicellulose xylan, which has immunomodulatory effects, has been combined with chitosan to form a composite hydrogel to improve the healing of bone fractures. This thermally responsive and injectable hydrogel, which is liquid at room temperature and gels at physiological temperature, improves the response of animal host tissue compared with similar pure chitosan hydrogels in tissue engineering models. The composite hydrogel was placed in a subcutaneous model where the composite hydrogel is replaced by host tissue within 1 week, much earlier than chitosan hydrogels. A tibia fracture model in mice showed that the composite encourages major remodeling of the fracture callus in less than 4 weeks. A non-union fracture model in rat femurs was used to demonstrate that the composite hydrogel allows bone regeneration and healing of defects that with no treatment are unhealed after 6 weeks. These results suggest that the xylan/chitosan composite hydrogel is a suitable bone graft substitute able to aid in the repair of large bone defects.

9.
Spine J ; 14(3): 445-54, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24360747

RESUMO

BACKGROUND CONTEXT: Although the use of mesenchymal stem cells (MSC) with scaffolds for bone repair has been considered an effective method, the interactions between implanted materials and bone tissues have not been fully elucidated. At some specific sites, such as the vertebral body (VB) of the spine, the process of bone repair with implanted biomaterials is rarely reported. Recently, adipose tissue was found to be an alternative source of MSC besides bone marrow. However, the strategy of using adipose-derived stromal (ADS) cells with bioactive scaffold for the repair of spinal bone defects has seldom been studied. PURPOSE: To use a sintered poly(lactide-co-glycolide) acid (PLGA) microspheres scaffold seeded with induced rat ADS cells to repair a bone defect of the VB in a rat model. STUDY DESIGN: Basic science and laboratory study. METHODS: A sintered porous microspheres scaffold was manufactured by PLGA. ADS cells were isolated from Fischer 344 rats and then induced by osteogenic medium with growth and differentiation factor 5 (GDF5) in vitro. Before implantation, cells were cultured with inductive media for 2 weeks as a monolayer situation and 1 more week on a PLGA scaffold as a three-dimensional structure. These assembled bioactive scaffolds then were implanted in lumbar VB bone defects in Fischer 344 rats. The ex vivo differentiation of the cells was confirmed by von Kossa staining and real-time polymerase chain reaction. The performance of cells on the scaffold was detected by scanning electron microscopy and (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay. In vivo bone formation was quantitatively measured by computed tomography study. And the effect of tissue repair was also evaluated by histological studies. RESULTS: Proliferation and differentiation of cells were confirmed before in vivo implantation. Quantification of bone formation in vivo through serial three-dimensional computed tomography images revealed that the VB implanted with GDF5-induced cells demonstrated more bone formation than the control groups. Besides the bone formation period that occurred between 2 and 4 weeks in all groups, a second bone formation period was found to occur only in the groups that received cells with previous induction in vitro. This second period of significant bone formation happened simultaneously with collapsing of the scaffolds. It was then demonstrated histologically that vascularization early in the process and cooperation between host bone and implanted cells accompanied by collapse of the scaffold may be the factors that influence bone formation. This study not only provides a therapeutic strategy of using biomaterial for bone repair in the spine, but also may lead to a technological method for studying the relationship between implanted stem cells and host tissue. CONCLUSIONS: Adipose-derived stromal cells maintained in culture on a scaffold and treated with osteogenic induction with growth factor ex vivo could be used to enhance bone repair in vivo.


Assuntos
Tecido Adiposo/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Doenças da Coluna Vertebral/cirurgia , Coluna Vertebral/cirurgia , Células Estromais/citologia , Alicerces Teciduais , Animais , Materiais Biocompatíveis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Fator 5 de Diferenciação de Crescimento/farmacologia , Ácido Láctico , Masculino , Microesferas , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Endogâmicos F344
10.
Spine J ; 13(1): 32-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23384881

RESUMO

BACKGROUND CONTEXT: Healthy mammalian cells in normal tissues are organized in complex three-dimensional (3D) networks that display nutrient and signaling gradients. Conventional techniques that grow cells in a 2D monolayer fail to reproduce the environment that is observed in vivo. In recent years, 3D culture systems have been used to mimic tumor microenvironments in cancer research and to emulate embryogenesis in stem cell cultures. However, there have been no studies exploring the ability for adipose-derived stromal (ADS) cells in a 3D culture system to undergo osteogenic differentiation. PURPOSE: To characterize and investigate the in vitro and in vivo potential for human ADS cells in a novel 3D culture system to undergo osteogenic differentiation. STUDY DESIGN: Basic science and laboratory study. METHODS: Human ADS cells were isolated and prepared as either a 2D monolayer or 3D multicellular aggregates (MAs). Multicellular aggregates were formed using the hanging droplet technique. Cells were treated in osteogenic medium in vitro, and cellular differentiation was investigated using gene expression, histology, and microCT at 1-, 2-, and 4-week time points. In vivo investigation involved creating a muscle pouch by developing the avascular muscular interval in the vastus lateralis of male athymic rats. Specimens were then pretreated with osteogenic medium and surgically implanted as (1) carrier (Matrigel) alone (control), (2) carrier with human ADS cells in monolayer, or (3) human ADS cells as MAs. In vivo evidence of osteogenic differentiation was evaluated with micro computed tomography and histologic sectioning at a 2-week time point. RESULTS: Human ADS cells cultured by the hanging droplet technique successfully formed MAs at the air-fluid interface. Adipose-derived stromal cells cultured in monolayer or as 3D MAs retain their ability to self-replicate and undergo multilineage differentiation as confirmed by increased runx2/Cbfa2, ALP, and OCN and increased matrix mineralization on histologic sectioning. Multicellular aggregate cells expressed increased differentiation potential and extracellular matrix production over the same human ADS cells cultured in monolayer. Furthermore, MAs reseeded onto monolayer retained their stem cell capabilities. When implanted in vivo, significantly greater bone volume and extracellular matrix were present in the implanted specimens of MAs confirmed on both microCT and histological sectioning. CONCLUSIONS: This is the first study to investigate the capability of human ADS cells in a 3D culture system to undergo osteogenic differentiation. The results confirm that MAs maintain their stem cell characteristics. Compared with analogous cells in monolayer culture, the human ADS cells as MAs exhibit elevated levels of osteogenic differentiation and increased matrix mineralization. Furthermore, the creation of uniform spheroids allows for improved handling and manipulation during transplantation. These findings strongly support the concept that 3D culture systems remain not only a viable option for stem cell culture but also possibly a more attractive alternative to traditional culture techniques to improve the osteogenic potential of human adipose stem cells.


Assuntos
Tecido Adiposo/citologia , Técnicas de Cultura de Células/métodos , Osteogênese/fisiologia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Células Estromais/citologia , Animais , Calcificação Fisiológica/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Masculino , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/cirurgia , Osteócitos/citologia , Ratos , Ratos Nus , Engenharia Tecidual/métodos , Tomografia Computadorizada por Raios X
11.
J Am Assoc Lab Anim Sci ; 50(2): 198-204, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21439213

RESUMO

Preventing and minimizing pain in laboratory animals is a basic tenet of biomedical research and is warranted for ethical, legal, and scientific reasons. Postoperative analgesia is an important facet of pain management. A sustained-release formulation of buprenorphine was tested in rats for analgesic efficacy and plasma concentration over a 72-h time period. Rats were injected subcutaneously with either 1.2 mg/kg sustained-release formulation (Bup-SR), 0.2 mL/kg buprenorphine HCl (Bup-HCl), or an equivalent volume of sustained-release vehicle and tested in a thermal nociception model or a surgical postoperative pain model. In both models, Bup-SR showed evidence of providing analgesia for 2 to 3 d. Thermal latency response in rats that received the sustained-release formulation increased 28.4% and 15.6% compared with baseline values on days 1 and 2, respectively. Rats with a unicortical tibial defect and treated with Bup-SR showed similar willingness to bear weight on the hindlimbs as did negative-control animals (no surgery), demonstrated by counting vertical raises; rats treated with Bup-HCl had significantly fewer vertical raises than did control rats for 5 d after surgery. Plasma concentrations of buprenorphine remained over 1 ng/mL for 72 h after a single dose of Bup-SR. Taken together, the results indicate that this formulation of buprenorphine may be a viable option for treating postsurgical pain in laboratory rats.


Assuntos
Analgesia/veterinária , Analgésicos Opioides/farmacologia , Buprenorfina/farmacologia , Medição da Dor/veterinária , Dor Pós-Operatória/veterinária , Ratos , Analgesia/métodos , Analgésicos Opioides/sangue , Animais , Buprenorfina/sangue , Preparações de Ação Retardada/farmacologia , Combinação de Medicamentos , Injeções Subcutâneas/veterinária , Masculino , Dor Pós-Operatória/tratamento farmacológico , Ratos Sprague-Dawley
12.
Spine (Phila Pa 1976) ; 36(7): 497-504, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21422981

RESUMO

STUDY DESIGN: In vivo experiments to develop a rat spine single metastasis model by using human breast cancer cells. OBJECTIVE: To study the survival and tumorigenesis of the human breast cancer cells after transplantation to vertebral body (VB) by intraosseous injection as a model for therapeutic studies of spine metastatic tumor. SUMMARY OF BACKGROUND DATA: VBs are the most common bones involved in the metastases of breast cancer. To develop experimental therapeutics requires an appropriate animal model. Moreover, it is also important to establish accurate and sensitive detection methods for the evaluation. METHODS: MDA-MB-231 human breast cancer cells were injected into 3-week-old female athymic rats. The tumorigenesis was assayed with quantitative in vivo bioluminescence (IVIS), microcomputed tomography (micro-CT), quantitative CT (qCT), micro position emission tomography (micro-PET), and histologic studies. RESULTS: A spine single metastasis model of human breast cancer was successfully developed in rats. The IVIS signal intensity from the cancer cells increased after 2 weeks. Signal from the tumor in spine can be detected by micro-PET at day 1. The signal intensity decreased after 1 week and then recovered and continually increased afterwards. Bone destruction was demonstrated in the qCT and micro-CT images. However, both qCT and micro-CT found that the bone density in the cancer cell-injected VB increased before the appearance of osteolysis. The growth of tumor and the reaction of bone in the VB were observed simultaneously by histology. CONCLUSION: A spine single metastasis model was developed by injection of human breast cancer cells into the VB of athymic rats. This is the first report of quantitative evaluation with micro-PET in a spine metastasis model. In addition, the detection of osteogenesis after the introduction of MDA-MB-231 cells in vivo is a novel observation.


Assuntos
Modelos Animais de Doenças , Neoplasias Mamárias Experimentais/patologia , Neoplasias da Medula Espinal/patologia , Neoplasias da Medula Espinal/secundário , Animais , Densidade Óssea/fisiologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Feminino , Humanos , Infusões Intraósseas , Ratos , Ratos Nus
13.
J Vis Exp ; (46)2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21178970

RESUMO

Two novel synthetic peptides accelerate bone formation and can be delivered using a collagen matrix. The aim of this study was to investigate the effects on bone repair in a unicortical defect model. Treatment of mesenchymal cells produced an increase in alkaline phosphatase activity, showed nodule formation by the cells, and increased the expression of genes for runx2, osterix, bone sialoprotein, and osteocalcin. A collagen sponge soaked with peptide promoted repair of bone defects, whereas the control was less effective. The results from this study demonstrated that mesenchymal cells treated with peptide in vitro differentiate towards osteogenesis, and, that peptides delivered in vivo using a collagen sponge promote the repair of unicortical defects.


Assuntos
Doenças Ósseas/tratamento farmacológico , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Biblioteca de Peptídeos , Peptídeos/farmacologia , Animais , Doenças Ósseas/genética , Doenças Ósseas/patologia , Regeneração Óssea/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Masculino , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Osteogênese/genética , Ratos , Ratos Endogâmicos F344
14.
Bone ; 47(2): 197-204, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20580872

RESUMO

Bone defects in vertebral bodies (VB) usually occur after the reduction of fractures or are caused by bone disease. Besides the treatment of original disease, repair of the bone defect can restore the structure of VB and improve stabilization of the spine to protect the spinal cord nerves. To aid studies of the efficacy of bioengineering techniques for repair of VB, we developed a rat model with a critical size bone defect in VB. Air-motivated burrs were used to create two sizes of bone defect (2 x 3 x 1.5 mm; 2 x 3 x 3 mm) in the anterior part of VB in 6-month-old Fischer 344 rats. Quantitative CT analyses and histological assays demonstrated that neither defects self-repaired by 8 weeks post surgery. Moreover, the tendency of bone formation was monitored in the same animal by serial CT image evaluations, allowing us to demonstrate that there was significant bone growth during the 4- to 6-week period after the creation of the bone defect. We then implanted sintered poly(lactic-co-glycolic acid) (PLGA) microsphere scaffolds loaded with Matrigel with or without recombinant human bone morphogenetic protein 2 (rhBMP2; 2.0 microg rhBMP2/10 microL Matrigel/scaffold) into the bone defect (2 x 3 x 3 mm) in the VB. Bone formation was detected by quantitative analyses of serial CT images, which demonstrated bone growth in rats that received the rhBMP2 implant, in both surrounding areas and inside area of the scaffold. In addition to a rapid increase within 2 weeks of the operation, another significant bone formation period was found between 4 and 8 weeks after the implantation. By contrast, the control group that received the implant without rhBMP2 did not show similar bone formation tendencies. The results of CT analyses were confirmed by histological studies. This study suggests that a critical size bone defect of the anterior VB can be developed in a rat model. Characterization of this model demonstrated that 4 to 6 weeks after creation of the defect was a significant bone growth period for VB bone repair in rats. This animal model has further utility for the study of different biomaterials for VB bone repair. Implantation of a bioactive PLGA scaffold carrying rhBMP2 allowed more successful repair of the VB defect. Although further characterization studies are needed, the bioactive PLGA scaffold developed in this study will likely adapt easily to other in vivo osteogenesis applications.


Assuntos
Materiais Biocompatíveis/farmacologia , Ácido Láctico/farmacologia , Ácido Poliglicólico/farmacologia , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/patologia , Alicerces Teciduais/química , Cicatrização/efeitos dos fármacos , Animais , Colágeno/metabolismo , Modelos Animais de Doenças , Combinação de Medicamentos , Laminina/metabolismo , Masculino , Microscopia Eletrônica de Varredura , Microesferas , Osteogênese/efeitos dos fármacos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Proteoglicanas/metabolismo , Ratos , Ratos Endogâmicos F344 , Reprodutibilidade dos Testes , Coluna Vertebral/diagnóstico por imagem , Tomografia Computadorizada por Raios X
15.
Spine J ; 10(1): 32-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19926342

RESUMO

BACKGROUND CONTEXT: The therapeutic strategies that have thus far been used for the treatment of intervertebral disc degeneration (IDD) have focused on relieving the symptoms, although reversal of the degeneration remains an important challenge for the effective treatment of IDD. Growth and differentiation factor-5 (GDF5), of which deficiency leads to early disc degeneration changes, has the potential to increase proliferation of disc cells and expression of extracellular matrix proteins. PURPOSE: The purpose of the study was to develop a lumbar disc degeneration model in mice and determine the effect of adenoviral GDF5 gene therapy. STUDY DESIGN: The study design was to compare the degeneration changes of discs punctured by different-size needles to develop a mice lumbar disc degeneration model and to evaluate the effects of in vivo gene therapy for the mice disc degeneration model by an adenoviral vector carrying GDF5 gene. METHODS: A lumbar disc degeneration model was developed by needle punctures to the discs in Balb/c mice. Afterward, a gene therapy treatment to disc degeneration was evaluated. Two of the mice lumbar discs were randomly chosen to be punctured by a 30-gauge needle and then injected with adenovirus that had been engineered to express either the luciferase gene (Ad-Luc) or the GDF5 gene (Ad-GDF5). Animals were analyzed by bioluminescent imaging, radiographic, and magnetic resonance imaging (MRI) scanning, then sacrificed at 1, 2, 4, or 8 weeks after operation, and subjected to histological and biochemical assays. RESULTS: By the detection of T2-weighted MRI scanning and histological study, the degeneration was found in all of the discs punctured by different-size needles. But the development of the degeneration in the discs injured by the 30-gauge needle was more reliable and moderate compared with that in other groups. The detection of luciferase activity by bioluminescent imaging revealed that adenovirus survived and the introduced genes were expressed over 6 weeks after injection. There were no T2-weighted MRI signals in the mice injected with either Ad-Luc or Ad-GDF5 up to 4 weeks after operation. At 6 and 8 weeks, T2-weighted signals were detected in the Ad-GDF5 group but none in the Ad-Luc control group. The percent disc height index (%DHI) was significantly decreased (approximately 20%) by 1 week after injury in both groups, indicating the development of disc degeneration. At 2 weeks, the %DHI in the mice injected with Ad-GDF5 increased significantly compared with that of the mice injected with Ad-Luc; the increase was sustained for the rest of the experiment period. The disc histology treated with Ad-GDF5 was improved compared with that in the control group. Glycosaminoglycan (GAG) levels were significantly decreased in the Ad-Luc injection group since 2 weeks after injury, and the DNA content had diminished by 4 weeks after the operation. In contrast, in the discs injected with Ad-GDF5, there was no decrease in the GAG and DNA levels after injury throughout the 8-week treatment period. CONCLUSIONS: Disc degeneration animal model can be developed by using needle puncture to the discs in mice. The adenovirus is an effective vehicle for gene delivery with rapid and prolonged expression of target protein and resulting improvement in markers of disc degeneration. Ad-GDF5 gene therapy could restore the functions of injured discs and has the potential to be an effective treatment.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Fator 5 de Diferenciação de Crescimento/genética , Degeneração do Disco Intervertebral/terapia , Animais , Divisão Celular/fisiologia , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/fisiologia , Disco Intervertebral/patologia , Disco Intervertebral/fisiologia , Degeneração do Disco Intervertebral/genética , Degeneração do Disco Intervertebral/patologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Ferimentos Penetrantes Produzidos por Agulha
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA