Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2783: 349-365, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478246

RESUMO

It is critical that human adipose-derived stromal/stem cell (hASC) tissue engineering therapies possess appropriate mechanical properties in order to restore the function of the load-bearing tissues of the musculoskeletal system. In an effort to elucidate hASC response to mechanical stimulation and develop mechanically robust tissue-engineered constructs, recent research has utilized a variety of mechanical loading paradigms, including cyclic tensile strain, cyclic hydrostatic pressure, and mechanical unloading in simulated microgravity. This chapter will describe the methods for applying these mechanical stimuli to hASC to direct differentiation for functional tissue engineering of the musculoskeletal system.


Assuntos
Sistema Musculoesquelético , Ausência de Peso , Humanos , Engenharia Tecidual/métodos , Pressão Hidrostática , Diferenciação Celular , Células-Tronco , Células Cultivadas
2.
F S Sci ; 5(1): 58-68, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38145868

RESUMO

OBJECTIVE: To assess the in vivo biomechanical maturation of tissue-engineered neo-uteri that have previously supported live births in a rabbit model. DESIGN: Nonclinical animal study. SETTING: University-based research laboratory. ANIMALS: Eighteen adult female rabbits. INTERVENTION: Biodegradable poly-DL-lactide-co-glycolide-coated polyglycolic acid scaffolds seeded with autologous uterine-derived endometrial and myometrial cells. Nonseeded scaffolds and seeded, tissue-engineered neo-uteri were implanted into one uterine horn of rabbits for 1, 3, or 6 months, excised, and biomechanically assessed in comparison to native uterine tissue. MAIN OUTCOME MEASURES: Tensile stress-relaxation testing, strain-to-failure testing, and viscoelastic modeling. RESULTS: By evaluating the biomechanical data with several viscoelastic models, it was revealed that tissue-engineered uteri were more mechanically robust than nonseeded scaffolds. For example, the 10% instantaneous stress of the tissue-engineered neo-uteri was 2.1 times higher than the nonseeded scaffolds at the 1-month time point, 1.6 times higher at the 3-month time point, and 1.5 times higher at the 6-month time point. Additionally, as the duration of implantation increased, the engineered constructs became more mechanically robust (e.g., 10% instantaneous stress of the tissue-engineered neo-uteri increased from 22 kPa at 1 month to 42 kPa at 6 months). Compared with native tissue values, tissue-engineered neo-uteri achieved or surpassed native tissue values by the 6-month time point. CONCLUSION: The present study evaluated the mechanical characteristics of novel tissue-engineered neo-uteri that have previously been reported to support live births in the rabbit model. We demonstrate that the biomechanics of these implants closely resemble those of native tissue, giving further credence to their development as a clinical solution to uterine factor infertility.


Assuntos
Engenharia Tecidual , Tecidos Suporte , Humanos , Gravidez , Animais , Feminino , Coelhos , Ácido Poliglicólico , Nascido Vivo , Útero/cirurgia
3.
J Biomed Mater Res B Appl Biomater ; 109(12): 2246-2258, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34114736

RESUMO

There is a need for the development of effective treatments for focal articular cartilage injuries. We previously developed a multiphasic 3D-bioplotted osteochondral scaffold design that can drive site-specific tissue formation when seeded with adipose-derived stem cells (ASC). The objective of this study was to evaluate this scaffold in a large animal model. Osteochondral defects were generated in the trochlear groove of Yucatan minipigs and repaired with scaffolds that either contained or lacked an electrospun tidemark and were either unseeded or seeded with ASC. Implants were monitored via computed tomography (CT) over the course of 4 months of in vivo implantation and compared to both open lesions and autologous explants. ICRS II evaluation indicated that defects with ASC-seeded scaffolds had healing that most closely resembled the aulogous explant. Scaffold-facilitated subchondral bone repair mimicked the structure of native bone tissue, but cartilage matrix staining was not apparent within the scaffold. The open lesions had the highest volumetric infill detected using CT analysis (p < 0.05), but the repair tissue was largely disorganized. The acellular scaffold without a tidemark had significantly more volumetric filling than either the acellular or ASC seeded groups containing a tidemark (p < 0.05), suggesting that the tidemark limited cell infiltration into the cartilage portion of the scaffold. Overall, scaffold groups repaired the defect more successfully than an open lesion but achieved limited repair in the cartilage region. With further optimization, this approach holds potential to treat focal cartilage lesions in a highly personalized manner using a human patient's own ASC cells.


Assuntos
Cartilagem Articular , Engenharia Tecidual , Animais , Cartilagem Articular/lesões , Células-Tronco , Suínos , Porco Miniatura , Engenharia Tecidual/métodos , Tecidos Suporte/química
4.
Mol Ther ; 28(4): 1056-1067, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32109370

RESUMO

Pre-clinical and clinical studies have shown that engineered tumoricidal neural stem cells (tNSCs) are a promising treatment strategy for the aggressive brain cancer glioblastoma (GBM). Yet, stabilizing human tNSCs within the surgical cavity following GBM resection is a significant challenge. As a critical step toward advancing engineered human NSC therapy for GBM, we used a preclinical variant of the clinically utilized NSC line HB1.F3.CD and mouse models of human GBM resection/recurrence to identify a polymeric scaffold capable of maximizing the transplant, persistence, and tumor kill of NSC therapy for post-surgical GBM. Using kinetic bioluminescence imaging, we found that tNSCs delivered into the mouse surgical cavity wall by direct injection persisted only 3 days. We found that delivery of tNSCs into the cavity on nanofibrous electrospun poly-l-lactic acid scaffolds extended tNSC persistence to 8 days. Modifications to fiber surface coating, diameter, and morphology of the scaffold failed to significantly extend tNSC persistence in the cavity. In contrast, tNSCs delivered into the post-operative cavity on gelatin matrices (GEMs) persisted 8-fold longer as compared to direct injection. GEMs remained permissive to tumor-tropic homing, as tNSCs migrated off the scaffolds and into invasive tumor foci both in vitro and in vivo. To mirror envisioned human brain tumor trials, we engineered tNSCs to express the prodrug/enzyme thymidine kinase (tNSCstk) and transplanted the therapeutic cells in the post-operative cavity of mice bearing resected orthotopic patient-derived GBM xenografts. Following administration of the prodrug ganciclovir, residual tumor volumes in mice receiving GEM/tNSCs were reduced by 10-fold at day 35, and median survival was extended from 31 to 46 days. Taken together, these data begin to define design parameters for effective scaffold/tNSC composites and suggest a new approach to maximizing the efficacy of tNSC therapy in human patient trials.


Assuntos
Neoplasias Encefálicas/terapia , Ganciclovir/administração & dosagem , Glioblastoma/terapia , Células-Tronco Neurais/transplante , Timidina Quinase/metabolismo , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Terapia Combinada , Ganciclovir/farmacologia , Glioblastoma/patologia , Glioblastoma/cirurgia , Humanos , Medições Luminescentes , Camundongos , Células-Tronco Neurais/metabolismo , Poliésteres/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacologia , Tecidos Suporte/química , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Biomed Mater Res B Appl Biomater ; 108(5): 2017-2030, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31880408

RESUMO

Osteoarthritis is a degenerative joint disease that limits mobility of the affected joint due to the degradation of articular cartilage and subchondral bone. The limited regenerative capacity of cartilage presents significant challenges when attempting to repair or reverse the effects of cartilage degradation. Tissue engineered medical products are a promising alternative to treat osteochondral degeneration due to their potential to integrate into the patient's existing tissue. The goal of this study was to create a scaffold that would induce site-specific osteogenic and chondrogenic differentiation of human adipose-derived stem cells (hASC) to generate a full osteochondral implant. Scaffolds were fabricated using 3D-bioplotting of biodegradable polycraprolactone (PCL) with either ß-tricalcium phosphate (TCP) or decellularized bovine cartilage extracellular matrix (dECM) to drive site-specific hASC osteogenesis and chondrogenesis, respectively. PCL-dECM scaffolds demonstrated elevated matrix deposition and organization in scaffolds seeded with hASC as well as a reduction in collagen I gene expression. 3D-bioplotted PCL scaffolds with 20% TCP demonstrated elevated calcium deposition, endogenous alkaline phosphatase activity, and osteopontin gene expression. Osteochondral scaffolds comprised of hASC-seeded 3D-bioplotted PCL-TCP, electrospun PCL, and 3D-bioplotted PCL-dECM phases were evaluated and demonstrated site-specific osteochondral tissue characteristics. This technique holds great promise as cartilage morbidity is minimized since autologous cartilage harvest is not required, tissue rejection is minimized via use of an abundant and accessible source of autologous stem cells, and biofabrication techniques allow for a precise, customizable methodology to rapidly produce the scaffold.


Assuntos
Materiais Biocompatíveis/química , Condrogênese/fisiologia , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Poliésteres/química , Tecidos Suporte/química , Tecido Adiposo/metabolismo , Osso e Ossos , Fosfatos de Cálcio/química , Fosfatos de Cálcio/metabolismo , Cartilagem Articular/metabolismo , Diferenciação Celular , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Poliésteres/metabolismo , Impressão Tridimensional , Engenharia Tecidual
6.
PLoS One ; 14(10): e0223245, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31584963

RESUMO

Mechanical loading is essential for the maintenance of musculoskeletal homeostasis. Cartilage has been demonstrated to be highly mechanoresponsive, but the mechanisms by which chondrocytes respond to mechanical stimuli are not clearly understood. The goal of the study was to determine how LRP4, LRP5, and LRP6 within canonical Wnt-signaling are regulated in simulated microgravity and cyclic hydrostatic pressure, and to investigate the potential role of LRP 4/5/6 in cartilage degeneration. Rat chondrosacroma cell (RCS) pellets were stimulated using either cyclic hydrostatic pressure (1Hz, 7.5 MPa, 4hr/day) or simulated microgravity in a rotating wall vessel (RWV) bioreactor (11RPM, 24hr/day). LRP4/5/6 mRNA expression was assessed by RT-qPCR and LRP5 protein expression was determined by fluorescent immunostaining. To further evaluate our in vitro findings in vivo, mice were subjected to hindlimb suspension for 14 days and the femoral heads stained for LRP5 expression. We found that, in vitro, LRP4/5/6 mRNA expression is modulated in a time-dependent manner by mechanical stimulation. Additionally, LRP5 protein expression is upregulated in response to both simulated microgravity and cyclic hydrostatic pressure. LRP5 is also upregulated in vivo in the articular cartilage of hindlimb suspended mice. This is the first study to examine how LRP4/5/6, critical receptors within musculoskeletal biology, respond to mechanical stimulation. Further elucidation of this mechanism could provide significant clinical benefit for the identification of pharmaceutical targets for the maintenance of cartilage health.


Assuntos
Condrócitos/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Mecanotransdução Celular/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Cartilagem Articular/citologia , Diferenciação Celular , Linhagem Celular Tumoral , Elevação dos Membros Posteriores/fisiologia , Pressão Hidrostática , Masculino , Camundongos , Modelos Animais , Ratos , Estresse Mecânico , Regulação para Cima
7.
Sci Rep ; 9(1): 8009, 2019 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-31142808

RESUMO

Non-motile primary cilia are dynamic cellular sensory structures and are expressed in adipose-derived stem cells (ASCs). We have previously shown that primary cilia are involved in chemically-induced osteogenic differentiation of human ASC (hASCs) in vitro. Further, we have reported that 10% cyclic tensile strain (1 Hz, 4 hours/day) enhances hASC osteogenesis. We hypothesize that primary cilia respond to cyclic tensile strain in a lineage dependent manner and that their mechanosensitivity may regulate the dynamics of signaling pathways localized to the cilium. We found that hASC morphology, cilia length and cilia conformation varied in response to culture in complete growth, osteogenic differentiation, or adipogenic differentiation medium, with the longest cilia expressed in adipogenically differentiating cells. Further, we show that cyclic tensile strain both enhances osteogenic differentiation of hASCs while it suppresses adipogenic differentiation as evidenced by upregulation of RUNX2 gene expression and downregulation of PPARG and IGF-1, respectively. This study demonstrates that hASC primary cilia exhibit mechanosensitivity to cyclic tensile strain and lineage-dependent expression, which may in part regulate signaling pathways localized to the primary cilium during the differentiation process. We highlight the importance of the primary cilium structure in mechanosensing and lineage specification and surmise that this structure may be a novel target in manipulating hASC for in tissue engineering applications.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Mecanotransdução Celular/genética , Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Cílios/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Células-Tronco Mesenquimais/citologia , Resistência à Tração , Engenharia Tecidual
8.
Toxicol Sci ; 167(1): 145-156, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30203000

RESUMO

Multipotent mesenchymal stem cells (MSCs) maintain the ability to differentiate into adipogenic, chondrogenic, or osteogenic cell lineages. There is increasing concern that exposure to environmental agents such as aryl hydrocarbon receptor (AhR) ligands, may perturb the osteogenic pathways responsible for normal bone formation. The objective of the current study was to evaluate the potential of the prototypic AhR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) to disrupt osteogenic differentiation of human bone-derived MSCs (hBMSCs) in vitro. Primary hBMSCs from three donors were exposed to 10 nM TCDD and differentiation was interrogated using select histological, biochemical, and transcriptional markers of osteogenesis. Exposure to 10 nM TCDD resulted in an overall consistent attenuation of alkaline phosphatase (ALP) activity and matrix mineralization at terminal stages of differentiation in primary hBMSCs. At the transcriptional level, the transcriptional regulator DLX5 and additional osteogenic markers (ALP, OPN, and IBSP) displayed attenuated expression; conversely, FGF9 and FGF18 were consistently upregulated in each donor. Expression of stem cell potency markers SOX2, NANOG, and SALL4 decreased in the osteogenic controls, whereas expression in TCDD-treated cells resembled that of undifferentiated cells. Coexposure with the AhR antagonist GNF351 blocked TCDD-mediated attenuation of matrix mineralization, and either fully or partially rescued expression of genes associated with osteogenic regulation, extracellular matrix, and/or maintenance of multipotency. Thus, experimental evidence from this study suggests that AhR transactivation likely attenuates osteoblast differentiation in multipotent hBMSCs. This study also underscores the use of primary human MSCs to evaluate osteoinductive or osteotoxic potential of chemical and pharmacologic agents in vitro.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/agonistas , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/agonistas , Fator 9 de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/genética , Humanos , Ligantes , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , Regulação para Cima
9.
J Biomed Mater Res B Appl Biomater ; 107(4): 900-910, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30230684

RESUMO

In this study, we report an industrial-scale fabrication method of a multifunctional polymer composite as a scaffold material for bone tissue engineering. This study successfully demonstrated the potential of applying industrial polymer processing technologies to produce specially functionalized tissue engineering scaffolds. With the inclusion of a newly synthesized multifunctional additive, silver-doped-calcium phosphate (silver-CaP), the composite material exhibited excellent osteogenic inducibility of human adipose-derived stem cells (hASC) and satisfactory antibacterial efficacy against Escherichia coli and Staphylococcus aureus. Also, relative to previously reported methods of direct loading silver particles into polymeric materials, our composite exhibited significantly reduced silver associated cytotoxicity. The enhanced biocompatibility could be a significant advantage for materials to be used for regenerative medicine applications where clinical safety is a major consideration. The impact of different silver loading methodologies on hASC' osteogenic differentiation was also studied. Overall, the results of this study indicate a promising alternative approach to produce multifunctional scaffolds at industrial-scale with higher throughput, lower cost, and enhanced reproducibility. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 900-910, 2019.


Assuntos
Tecido Adiposo/metabolismo , Antibacterianos , Fosfatos de Cálcio , Escherichia coli/crescimento & desenvolvimento , Osteogênese/efeitos dos fármacos , Poliésteres , Prata , Staphylococcus aureus/crescimento & desenvolvimento , Células-Tronco/metabolismo , Tecido Adiposo/citologia , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Humanos , Poliésteres/síntese química , Poliésteres/química , Poliésteres/farmacologia , Prata/química , Prata/farmacologia , Células-Tronco/citologia , Tecidos Suporte/química
10.
J Tissue Eng Regen Med ; 12(12): 2277-2286, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30352487

RESUMO

Corin has been studied extensively within the vascular system and is known to regulate blood pressure. We have shown that corin is one of the most highly upregulated genes during osteogenic differentiation of human adipose-derived stem cells (hASCs). This study tested the hypothesis that, through modulation of angiogenic signalling pathways, corin is a critical regulator of osteogenic differentiation and endochondral ossification. In vitro, corin expression in hASC was suppressed via siRNA knockdown and vascular endothelial growth factor A (VEGF-A) expression was quantified via reverse transcription polymerase chain reaction. In vivo, a murine corin knockout model (female, 10 weeks) was used to determine the effect of corin deficiency on long bone development. Wild-type and corin knockout long bones were compared via haematoxylin and eosin staining to assess tissue characteristics and cellular organization, three-point bending to assess mechanical characteristics, and immunohistochemistry to visualize VEGF-A expression patterns. Corin knockdown significantly (p < 0.05) increased VEGF-A mRNA expression during osteogenic differentiation. In vivo, corin knockout reduced tibial growth plate thickness (p < 0.01) and severely diminished the hypertrophic region. Corin knockout femurs had significantly increased stiffness (p < 0.01) and maximum loads (p < 0.01) but reduced postyield deflections (p < 0.01). In corin knockout mice, VEGF-A expression was increased near the growth plate but was reduced throughout the tibial shaft and distal head of the tibiae. This is the first study to show that corin is a key regulator of bone development by modulation of VEGF-A expression. Further elucidation of this mechanism will aid in the development of optimized bone tissue engineering and regenerative medicine therapies.


Assuntos
Tecido Adiposo/metabolismo , Regulação da Expressão Gênica , Osteogênese , Serina Endopeptidases/biossíntese , Serina Endopeptidases/metabolismo , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Tecido Adiposo/citologia , Adulto , Animais , Feminino , Humanos , Camundongos , Camundongos Knockout , Serina Endopeptidases/genética , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/genética
11.
Methods Mol Biol ; 1773: 215-230, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29687393

RESUMO

It is critical that human adipose stem cell (hASC) tissue-engineering therapies possess appropriate mechanical properties in order to restore function of the load bearing tissues of the musculoskeletal system. In an effort to elucidate the hASC response to mechanical stimulation and develop mechanically robust tissue engineered constructs, recent research has utilized a variety of mechanical loading paradigms including cyclic tensile strain, cyclic hydrostatic pressure, and mechanical unloading in simulated microgravity. This chapter describes methods for applying these mechanical stimuli to hASC to direct differentiation for functional tissue engineering of the musculoskeletal system.


Assuntos
Condrogênese , Células-Tronco Mesenquimais/citologia , Osteogênese , Estresse Mecânico , Engenharia Tecidual/métodos , Simulação de Ausência de Peso , Tecido Adiposo/citologia , Diferenciação Celular , Humanos , Pressão Hidrostática , Sistema Musculoesquelético , Cultura Primária de Células , Resistência à Tração
12.
J Orthop Res ; 36(2): 605-619, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28817244

RESUMO

Advances in mechanobiology have evolved through insights from multiple disciplines including structural engineering, biomechanics, vascular biology, and orthopaedics. In this paper, we reviewed the impact of key reports related to the study of applied loads on tissues and cells and the resulting signal transduction pathways. We addressed how technology has helped advance the burgeoning field of mechanobiology (over 33,600 publications from 1970 to 2016). We analyzed the impact of critical ideas and then determined how these concepts influenced the mechanobiology field by looking at the citation frequency of these reports as well as tracking how the overall number of citations within the field changed over time. These data allowed us to understand how a key publication, idea, or technology guided or enabled the field. Initial observations of how forces acted on bone and soft tissues stimulated the development of computational solutions defining how forces affect tissue modeling and remodeling. Enabling technologies, such as cell and tissue stretching, compression, and shear stress devices, allowed more researchers to explore how deformation and fluid flow affect cells. Observation of the cell as a tensegrity structure and advanced methods to study genetic regulation in cells further advanced knowledge of specific mechanisms of mechanotransduction. The future of the field will involve developing gene and drug therapies to simulate or augment beneficial load regimens in patients and in mechanically conditioning organs for implantation. Here, we addressed a history of the field, but we limited our discussions to advances in musculoskeletal mechanobiology, primarily in bone, tendon, and ligament tissues. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:605-619, 2018.


Assuntos
Biofísica/história , Animais , Biofísica/métodos , História do Século XIX , História do Século XX , Humanos , Mecanotransdução Celular
13.
Tissue Eng Part C Methods ; 23(11): 795-803, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28750575

RESUMO

Surgical site infection (SSI) is the most common cause of surgical failure, increasing the risks of postoperative mortality and morbidity. Recently, it has been reported that the use of antimicrobial dressings at the incision site help with prevention of SSI. Despite the increased body of research on the development of different types of antimicrobial dressings for this application, to our knowledge, nobody has reported a reliable large animal model to evaluate the efficacy of developed materials in a preclinical SSI model. In this study, we developed a porcine full-thickness incision model to investigate SSI caused by methicillin-resistant Staphylococcus aureus (MRSA), the leading cause of SSI in the United States. Using this model, we then evaluated the efficacy of our newly developed silver releasing nanofibrous dressings for preventing and inhibiting MRSA infection. Our results confirmed the ease and practicality of a new porcine model as an in vivo platform for evaluation of biomaterials for SSI. Using this model, we found that our silver releasing scaffolds significantly reduced bacterial growth in wounds inoculated with MRSA relative to nontreated controls and to wounds treated with the gold standard, silver sulfadiazine, without causing inflammation at the wound site. Findings from this study confirm the potential of our silver-releasing nanofibrous scaffolds for treatment/prevention of SSI, and introduce a new porcine model for in vivo evaluation of additional SSI treatment approaches.


Assuntos
Infecção da Ferida Cirúrgica/tratamento farmacológico , Animais , Anti-Infecciosos/farmacologia , Anti-Infecciosos/uso terapêutico , Modelos Animais de Doenças , Feminino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Nanofibras/química , Nanofibras/ultraestrutura , Poliésteres/química , Porosidade , Infecção da Ferida Cirúrgica/microbiologia , Infecção da Ferida Cirúrgica/patologia , Sus scrofa , Tecidos Suporte/química
14.
Aerosp Med Hum Perform ; 88(4): 377-384, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28518000

RESUMO

BACKGROUND: Cartilage tissue engineering is a growing field due to the lack of regenerative capacity of native tissue. The use of bioreactors for cartilage tissue engineering is common, but the results are controversial. Some studies suggest that microgravity bioreactors are ideal for chondrogenesis, while others show that mimicking hydrostatic pressure is crucial for cartilage formation. A parallel study comparing the effects of loading and unloading on chondrogenesis has not been performed. METHODS: The goal of this study was to evaluate chondrogenesis of human adipose-derived stem cells (hASC) under two different mechanical stimuli relative to static culture: microgravity and cyclic hydrostatic pressure (CHP). Pellets of hASC were cultured for 14 d under simulated microgravity using a rotating wall vessel bioreactor or under CHP (7.5 MPa, 1 Hz, 4 h · d-1) using a hydrostatic pressure vessel. RESULTS: We found that CHP increased mRNA expression of Aggrecan, Sox9, and Collagen II, caused a threefold increase in sulfated glycosaminoglycan production, and resulted in stronger vimentin staining intensity and organization relative to microgravity. In addition, Wnt-signaling patterns were altered in a manner that suggests that simulated microgravity decreases chondrogenic differentiation when compared to CHP. DISCUSSION: Our goal was to compare chondrogenic differentiation of hASC using a microgravity bioreactor and a hydrostatic pressure vessel, two commonly used bioreactors in cartilage tissue engineering. Our results indicate that CHP promotes hASC chondrogenesis and that microgravity may inhibit hASC chondrogenesis. Our findings further suggest that cartilage formation and regeneration might be compromised in space due to the lack of mechanical loading.Mellor LF, Steward AJ, Nordberg RC, Taylor MA, Loboa EG. Comparison of simulated microgravity and hydrostatic pressure for chondrogenesis of hASC. Aerosp Med Hum Perform. 2017; 88(4):377-384.


Assuntos
Condrogênese/fisiologia , Pressão Hidrostática , Células-Tronco/citologia , Engenharia Tecidual , Simulação de Ausência de Peso , Reatores Biológicos , Técnicas de Cultura de Células , Diferenciação Celular , Humanos , Estimulação Física , Suporte de Carga
15.
ACS Appl Mater Interfaces ; 9(25): 21105-21115, 2017 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-28540723

RESUMO

In this study, we report a high-throughput fabrication method at industrial pilot scale to produce a silver-nanoparticles-doped nanoclay-polylactic acid composite with a novel synergistic antibacterial effect. The obtained nanocomposite has a significantly lower affinity for bacterial adhesion, allowing the loading amount of silver nanoparticles to be tremendously reduced while maintaining satisfactory antibacterial efficacy at the material interface. This is a great advantage for many antibacterial applications in which cost is a consideration. Furthermore, unlike previously reported methods that require additional chemical reduction processes to produce the silver-nanoparticles-doped nanoclay, an in situ preparation method was developed in which silver nanoparticles were created simultaneously during the composite fabrication process by thermal reduction. This is the first report to show that altered material surface submicron structures created with the loading of nanoclay enables the creation of a nanocomposite with significantly lower affinity for bacterial adhesion. This study provides a promising scalable approach to produce antibacterial polymeric products with minimal changes to industry standard equipment, fabrication processes, or raw material input cost.


Assuntos
Nanopartículas Metálicas , Antibacterianos , Nanocompostos , Polímeros , Prata
16.
Biomed Res Int ; 2017: 6956794, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28536700

RESUMO

Electrospun scaffolds provide a dense framework of nanofibers with pore sizes and fiber diameters that closely resemble the architecture of native extracellular matrix. However, it generates limited three-dimensional structures of relevant physiological thicknesses. 3D printing allows digitally controlled fabrication of three-dimensional single/multimaterial constructs with precisely ordered fiber and pore architecture in a single build. However, this approach generally lacks the ability to achieve submicron resolution features to mimic native tissue. The goal of this study was to fabricate and evaluate 3D printed, electrospun, and combination of 3D printed/electrospun scaffolds to mimic the native architecture of heterogeneous tissue. We assessed their ability to support viability and proliferation of human adipose derived stem cells (hASC). Cells had increased proliferation and high viability over 21 days on all scaffolds. We further tested implantation of stacked-electrospun scaffold versus combined electrospun/3D scaffold on a cadaveric pig knee model and found that stacked-electrospun scaffold easily delaminated during implantation while the combined scaffold was easier to implant. Our approach combining these two commonly used scaffold fabrication technologies allows for the creation of a scaffold with more close resemblance to heterogeneous tissue architecture, holding great potential for tissue engineering and regenerative medicine applications of osteochondral tissue and other heterogeneous tissues.


Assuntos
Impressão Tridimensional , Engenharia Tecidual/métodos , Tecidos Suporte/química , Animais , Células Cultivadas , Matriz Extracelular/química , Humanos , Nanofibras/química , Nanofibras/uso terapêutico , Poliésteres/química , Poliésteres/uso terapêutico , Porosidade , Suínos
17.
Stem Cells Transl Med ; 6(2): 502-511, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28191763

RESUMO

Human adipose stem cells (hASCs) are an attractive cell source for bone tissue engineering applications. However, a critical issue to be addressed before widespread hASC clinical translation is the dramatic variability in proliferative capacity and osteogenic potential among hASCs isolated from different donors. The goal of this study was to test our hypothesis that electrical cell-substrate impedance spectroscopy (ECIS) could track complex bioimpedance patterns of hASCs throughout proliferation and osteogenic differentiation to better understand and predict variability among hASC populations. Superlots composed of hASCs from young (aged 24-36 years), middle-aged (aged 48-55 years), and elderly (aged 60-81 years) donors were seeded on gold electrode arrays. Complex impedance measurements were taken throughout proliferation and osteogenic differentiation. During osteogenic differentiation, four impedance phases were identified: increase, primary stabilization, drop phase, and secondary stabilization. Matrix deposition was first observed 48-96 hours after the impedance maximum, indicating, for the first time, that ECIS can identify morphological changes that correspond to late-stage osteogenic differentiation. The impedance maximum was observed at day 10.0 in young, day 6.1 in middle-aged, and day 1.3 in elderly hASCs, suggesting that hASCs from younger donors require a longer time to differentiate than do hASCs from older donors, but young hASCs proliferated more and accreted more calcium long-term. This is the first study to use ECIS to predict osteogenic potential of multiple hASC populations and to show that donor age may temporally control onset of osteogenesis. These findings could be critical for development of patient-specific bone tissue engineering and regenerative medicine therapies. Stem Cells Translational Medicine 2017;6:502-511.


Assuntos
Tecido Adiposo/citologia , Osso e Ossos/citologia , Diferenciação Celular , Proliferação de Células , Espectroscopia Dielétrica/métodos , Osteogênese , Células-Tronco/fisiologia , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Impedância Elétrica , Humanos , Pessoa de Meia-Idade , Fenótipo , Células-Tronco/metabolismo , Fatores de Tempo , Adulto Jovem
18.
FASEB J ; 31(1): 346-355, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27825103

RESUMO

In this study, we report for the first time that the primary cilium acts as a crucial sensor for electrical field stimulation (EFS)-enhanced osteogenic response in osteoprogenitor cells. In addition, primary cilia seem to functionally modulate effects of EFS-induced cellular calcium oscillations. Primary cilia are organelles that have recently been implicated to play a crucial sensor role for many mechanical and chemical stimuli on stem cells. Here, we investigate the role of primary cilia in EFS-enhanced osteogenic response of human adipose-derived stem cells (hASCs) by knocking down 2 primary cilia structural proteins, polycystin-1 and intraflagellar protein-88. Our results indicate that structurally integrated primary cilia are required for detection of electrical field signals in hASCs. Furthermore, by measuring changes of cytoplasmic calcium concentration in hASCs during EFS, our findings also suggest that primary cilia may potentially function as a crucial calcium-signaling nexus in hASCs during EFS.-Cai, S., Bodle, J. C., Mathieu, P. S., Amos, A., Hamouda, M., Bernacki, S., McCarty, G., Loboa, E. G. Primary cilia are sensors of electrical field stimulation to induce osteogenesis of human adipose-derived stem cells.


Assuntos
Tecido Adiposo/citologia , Cílios/fisiologia , Estimulação Elétrica , Osteogênese/fisiologia , Células-Tronco/fisiologia , Biomarcadores , Cálcio/metabolismo , Sobrevivência Celular , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Humanos , Interferência de RNA , RNA Interferente Pequeno
19.
J Orthop Surg Res ; 11(1): 132, 2016 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-27793202

RESUMO

BACKGROUND: The meniscus plays a crucial role in knee joint stability, load transmission, and stress distribution. Meniscal tears are the most common reported knee injuries, and the current standard treatment for meniscal deficiency is meniscal allograft transplantation. A major limitation of this approach is that meniscal allografts do not have the capacity to remodel and maintain tissue homeostasis due to a lack of cellular infiltration. The purpose of this study was to provide a new method for enhanced cellular infiltration in meniscal allografts. METHODS: Twenty medial menisci were collected from cadaveric human sources and split into five experimental groups: (1) control native menisci, (2) decellularized menisci, (3) decellularized menisci seeded with human adipose-derived stem cells (hASC), (4) decellularized needle-punched menisci, and (5) decellularized needle-punched menisci seeded with hASC. All experimental allografts were decellularized using a combined method with trypsin EDTA and peracetic acid. Needle punching (1-mm spacing, 28 G microneedle) was utilized to improve porosity of the allograft. Samples were recellularized with hASC at a density of 250 k/g of tissue. After 28 days of in vitro culture, menisci were analyzed for mechanical, biochemical, and histological characteristics. RESULTS: Menisci maintained structural integrity and material properties (compressive equilibrium and dynamic moduli) throughout preparations. Increased DNA content was observed in the needle-punched menisci but not in the samples without needle punching. Histology confirmed these results, showing enhanced cellular infiltration in needle-punched samples. CONCLUSIONS: The enhanced infiltration achieved in this study could help meniscal allografts better remodel post-surgery. The integration of autologous adipose-derived stem cells could improve long-term efficacy of meniscal transplantation procedures by helping to maintain the meniscus in vivo.


Assuntos
Tecido Adiposo/citologia , Aloenxertos/citologia , Menisco/citologia , Agulhas , Transplante de Células-Tronco/métodos , Tecido Adiposo/fisiologia , Tecido Adiposo/transplante , Adulto , Aloenxertos/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Humanos , Masculino , Menisco/fisiologia , Menisco/transplante , Pessoa de Meia-Idade , Células-Tronco/fisiologia , Transplante Homólogo/métodos
20.
Tissue Eng Part A ; 22(21-22): 1258-1263, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27676280

RESUMO

Bone infection, also called osteomyelitis, can result when bacteria invade a bone. Treatment of osteomyelitis usually requires surgical debridement and prolonged antimicrobial therapy. The rising incidence of infection with multidrug-resistant bacteria, in particular methicillin-resistant staphylococcus aureus (MRSA), however, limits the antimicrobial treatment options available. Silver is well known for its antimicrobial properties and is highly toxic to a wide range of microorganisms. We previously reported our development of biocompatible, biodegradable, nanofibrous scaffolds that released silver ions in a controlled manner. The objective of this study was to determine the efficacy of these scaffolds in treating or preventing osteomyelitis. To achieve this objective, antimicrobial efficacy was determined using a 3D coculture system of human adipose-derived stem cells (hASC) and MRSA. Human ASC were seeded on the scaffolds and induced to undergo osteogenic differentiation in both the absence and presence of MRSA. Our results indicated that the silver ion-releasing scaffolds not only inhibited biofilm formation, but also supported osteogenesis of hASC. Our findings suggest that these biocompatible, degradable, silver ion-releasing scaffolds can be used at an infection site to treat osteomyelitis and/or to coat bone implants as a preventative measure against infection postsurgery.


Assuntos
Tecido Adiposo/metabolismo , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Osteomielite/prevenção & controle , Prata , Células-Tronco/metabolismo , Tecidos Suporte/química , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cocultura , Humanos , Osteogênese/efeitos dos fármacos , Prata/química , Prata/farmacocinética , Prata/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...