Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Hand (N Y) ; 16(6): 770-775, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-31690117

RESUMEN

Background: The gate control theory asserts that non-painful stimuli can block pain perception. The ShotBlocker™ device is a plastic disk with blunt projections that rests on the skin, and we hypothesize that it will reduce pain during hand injections. Methods: This is a prospective randomized trial of 117 patients undergoing injections for common hand conditions. Patients were randomized into 3 groups: device, placebo (device with projections removed), and control. Patients recorded on an analog pain scale the pain severity of the injection, as well as their most recent tetanus shot. A normalized pain score was obtained from the difference between the injection and tetanus shot pain scores. The mean non-normalized and normalized scores for each treatment group were compared to the control group using the Wilcoxon signed rank test. Results: There were 91 women and 26 men. Common diagnoses included trigger finger (n = 53), DeQuervain's tendonitis (n = 33), and basal joint arthritis (n = 22). The groups did not differ significantly in age, gender, or diagnosis. Mean pain score in the device group was 5.2 out of 10, and it was 5.7 for the control group. The normalized pain score in the device group was significantly lower than the control group. Normalized and non-normalized pain scores for the placebo group were not significantly lower than the control group. Conclusions: The shot blocking device effectively reduced pain of injection versus controls when pain scores were normalized for pain tolerance. The modified device did not reduce the pain of injection, suggesting that gate control is the mechanism of action.


Asunto(s)
Mano , Dolor , Femenino , Mano/cirugía , Humanos , Inyecciones , Masculino , Dimensión del Dolor , Estudios Prospectivos
2.
Cytotherapy ; 19(12): 1491-1500, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28917626

RESUMEN

BACKGROUND AIMS: Regenerative medicine employs human mesenchymal stromal cells (MSCs) for their multi-lineage plasticity and their pro-regenerative cytokine secretome. Adipose-derived mesenchymal stromal cells (ASCs) are concentrated in fat tissue, and the ease of harvest via liposuction makes them a particularly interesting cell source. However, there are various liposuction methods, and few have been assessed regarding their impact on ASC functionality. Here we study the impact of the two most popular ultrasound-assisted liposuction (UAL) devices currently in clinical use, VASER (Solta Medical) and Lysonix 3000 (Mentor) on ASCs. METHODS: After lipoaspirate harvest and processing, we sorted for ASCs using fluorescent-assisted cell sorting based on an established surface marker profile (CD34+CD31-CD45-). ASC yield, viability, osteogenic and adipogenic differentiation capacity and in vivo regenerative performance were assessed. RESULTS: Both UAL samples demonstrated equivalent ASC yield and viability. VASER UAL ASCs showed higher osteogenic and adipogenic marker expression, but a comparable differentiation capacity was observed. Soft tissue healing and neovascularization were significantly enhanced via both UAL-derived ASCs in vivo, and there was no significant difference between the cell therapy groups. CONCLUSIONS: Taken together, our data suggest that UAL allows safe and efficient harvesting of the mesenchymal stromal cellular fraction of adipose tissue and that cells harvested via this approach are suitable for cell therapy and tissue engineering applications.


Asunto(s)
Tejido Adiposo/citología , Lipectomía/instrumentación , Lipectomía/métodos , Células del Estroma/citología , Ultrasonografía/métodos , Adipocitos/citología , Adipogénesis , Tejido Adiposo/diagnóstico por imagen , Adulto , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Femenino , Citometría de Flujo/métodos , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones Desnudos , Persona de Mediana Edad , Osteogénesis , Regeneración , Cicatrización de Heridas
3.
Plast Reconstr Surg ; 140(3): 517-524, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28574947

RESUMEN

BACKGROUND: Because of the abundance and biocompatibility of fat, lipotransfer has become an attractive method for treating soft-tissue deficits. However, it is limited by unpredictable graft survival and retention. Currently, little is known about the viscoelastic properties of fat after various injection methods. Here, the authors assess the effects of cannula diameter, length, and shape on the viscoelastic properties, structure, and retention of fat. METHODS: Human lipoaspirate was harvested using suction-assisted liposuction and prepared for grafting. A syringe pump was used to inject fat at a controlled flow rate through cannulas of varying gauges, lengths, and shapes. Processed samples were tested in triplicate on an oscillatory rheometer to measure their viscoelastic properties. Fat grafts from each group were placed into the scalps of immunocompromised mice. After 8 weeks, graft retention was measured using micro-computed tomography and grafts were explanted for histologic analysis. RESULTS: Lipoaspirate injected through narrower, longer, and bent cannulas exhibited more shear thinning with diminished quality. The storage modulus (G') of fat processed with 18-gauge cannulas was significantly lower than when processed with 14-gauge or larger cannulas, which also corresponded with inferior in vivo histologic structure. Similarly, the longer cannula group had a significantly lower storage modulus than the shorter cannula, and was associated with decreased graft retention. CONCLUSIONS: Discrete modifications in the methods used for fat placement can have a significant impact on immediate graft integrity, and ultimately on graft survival and quality. Respecting these biomechanical influences during the placement phase of lipotransfer may allow surgeons to optimize outcomes. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Asunto(s)
Tejido Adiposo/fisiología , Tejido Adiposo/trasplante , Catéteres , Supervivencia de Injerto/fisiología , Trasplante de Tejidos/métodos , Adipocitos/trasplante , Animales , Modelos Animales de Enfermedad , Diseño de Equipo , Humanos , Ratones , Trasplante Autólogo , Microtomografía por Rayos X
4.
Stem Cells Transl Med ; 6(1): 151-160, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28170185

RESUMEN

Clinical translation of cell-based strategies for tissue regeneration remains challenging because survival of implanted cells within hostile, hypoxic wound environments is uncertain. Overexpression of B-cell lymphoma 2 (Bcl-2) has been shown to inhibit apoptosis in implanted cells. The present study describes an "off the shelf" prefabricated scaffold integrated with magnetic nanoparticles (MNPs) used to upregulate Bcl-2 expression in implanted adipose-derived stromal cells for bone regeneration. Iron oxide cores were sequentially coated with branched polyethyleneimine, minicircle plasmid encoding green fluorescent protein and Bcl-2, and poly-ß-amino ester. Through in vitro assays, increased osteogenic potential and biological resilience were demonstrated in the magnetofected group over control and nucleofected groups. Similarly, our in vivo calvarial defect study showed that magnetofection had an efficiency rate of 30%, which in turn resulted in significantly more healing compared with control group and nucleofected group. Our novel, prefabricated MNP-integrated scaffold allows for in situ postimplant temporospatial control of cell transfection to augment bone regeneration. Stem Cells Translational Medicine 2017;6:151-160.


Asunto(s)
Regeneración Ósea , Nanopartículas de Magnetita/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Regulación hacia Arriba , Tejido Adiposo/citología , Adulto , Animales , Supervivencia Celular , Regulación de la Expresión Génica , Humanos , Campos Magnéticos , Masculino , Ratones , Persona de Mediana Edad , Osteogénesis/genética , Células del Estroma/citología , Células del Estroma/trasplante , Andamios del Tejido/química
5.
Plast Reconstr Surg ; 138(6): 983e-996e, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27537222

RESUMEN

BACKGROUND: The authors have developed a novel protocol for isolating adipose-derived stem cells from human lipoaspirate. In this study, they compare their new method to a previously published standard protocol. METHODS: Human adipose-derived stem cell isolation was performed using two methods to compare cell yield, cell viability, cell proliferation, and regenerative potential. The new and conventional isolation methods differ in two key areas: the collagenase digestion buffer constituents and the use of an orbital shaker. The osteogenic and adipogenic potential of adipose-derived stem cells isolated using both protocols was assessed in vitro, and gene expression analysis was performed. To assess the ability of the isolated cells to generate bone in vivo, the authors created critical-size calvarial defects in mice, which were treated with adipose-derived stem cells loaded onto hydroxyapatite-coated poly(lactic-co-glycolic acid) scaffolds. To test the ability of the isolated cells to enhance adipogenesis, the cells were added to lipoaspirate and placed beneath the scalp of immunocompromised mice. Fat graft volume retention was subsequently assessed by serial computed tomographic volumetric scanning. RESULTS: The new method resulted in a 10-fold increased yield of adipose-derived stem cells compared with the conventional method. Cells harvested using the new method demonstrated significantly increased cell viability and proliferation in vitro (p < 0.05). New method cells also demonstrated significantly enhanced osteogenic and adipogenic differentiation capacity in vitro (p < 0.05) in comparison with the conventional method cells. Both cell groups demonstrated equivalent osteogenic and adipogenic regenerative potential in mice. CONCLUSIONS: The authors have developed a protocol that maximizes the yield of adipose-derived stem cells derived from lipoaspirate. The new method cells have increased osteogenic and adipogenic potential in vitro and are not inferior to conventional method cells in terms of their ability to generate bone and fat in vivo. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Asunto(s)
Separación Celular/métodos , Células Madre Mesenquimatosas , Grasa Subcutánea/citología , Adipogénesis , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Femenino , Humanos , Lipectomía , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Ratones , Osteogénesis , Ingeniería de Tejidos
6.
J Transl Med ; 14(1): 126, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-27153799

RESUMEN

BACKGROUND: Adipose-derived stem cells (ASCs) have been identified as a population of multipotent cells with promising applications in tissue engineering and regenerative medicine. ASCs are abundant in fat tissue, which can be safely harvested through the minimally invasive procedure of liposuction. However, there exist a variety of different harvesting methods, with unclear impact on ASC regenerative potential. The aim of this study was thus to compare the functionality of ASCs derived from the common technique of suction-assisted lipoaspiration (SAL) versus resection. METHODS: Human adipose tissue was obtained from paired abdominoplasty and SAL samples from three female donors, and was processed to isolate the stromal vascular fraction. Fluorescence-activated cell sorting was used to determine ASC yield, and cell viability was assayed. Adipogenic and osteogenic differentiation capacity were assessed in vitro using phenotypic staining and quantification of gene expression. Finally, ASCs were applied in an in vivo model of tissue repair to evaluate their regenerative potential. RESULTS: SAL specimens provided significantly fewer ASCs when compared to excised fat tissue, however, with equivalent viability. SAL-derived ASCs demonstrated greater expression of the adipogenic markers FABP-4 and LPL, although this did not result in a difference in adipogenic differentiation. There were no differences detected in osteogenic differentiation capacity as measured by alkaline phosphatase, mineralization or osteogenic gene expression. Both SAL- and resection-derived ASCs enhanced significantly cutaneous healing and vascularization in vivo, with no significant difference between the two groups. CONCLUSION: SAL provides viable ASCs with full capacity for multi-lineage differentiation and tissue regeneration, and is an effective method of obtaining ASCs for cell-based therapies.


Asunto(s)
Tejido Adiposo/citología , Lipectomía/métodos , Regeneración , Células Madre/citología , Abdominoplastia , Adipogénesis , Adulto , Animales , Recuento de Células , Diferenciación Celular , Linaje de la Célula , Supervivencia Celular , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neovascularización Fisiológica , Osteogénesis , Succión , Cicatrización de Heridas
7.
J Biomed Mater Res A ; 104(8): 2099-107, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27059085

RESUMEN

Scaffold-mediated gene delivery holds great promise for tissue regeneration. However, previous attempts to induce bone regeneration using scaffold-mediated non-viral gene delivery rarely resulted in satisfactory healing. We report a novel platform with sustained release of minicircle DNA (MC) from PLGA scaffolds to accelerate bone repair. MC was encapsulated inside PLGA scaffolds using supercritical CO2 , which showed prolonged release of MC. Skull-derived osteoblasts transfected with BMP-2 MC in vitro result in higher osteocalcin gene expression and mineralized bone formation. When implanted in a critical-size mouse calvarial defect, scaffolds containing luciferase MC lead to robust in situ protein production up to at least 60 days. Scaffold-mediated BMP-2 MC delivery leads to substantially accelerated bone repair as early as two weeks, which continues to progress over 12 weeks. This platform represents an efficient, long-term nonviral gene delivery system, and may be applicable for enhancing repair of a broad range of tissues types. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2099-2107, 2016.


Asunto(s)
Proteína Morfogenética Ósea 2/administración & dosificación , ADN Circular/metabolismo , Técnicas de Transferencia de Gen , Cráneo/patología , Andamios del Tejido/química , Cicatrización de Heridas , Animales , Proteína Morfogenética Ósea 2/metabolismo , Luciferasas/metabolismo , Ratones , Ratones Desnudos , Osteogénesis , Ácido Poliglicólico/química , Regulación hacia Arriba , Microtomografía por Rayos X
8.
J Vis Exp ; (107)2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26780559

RESUMEN

Fibroblasts are the principle cell type responsible for secreting extracellular matrix and are a critical component of many organs and tissues. Fibroblast physiology and pathology underlie a spectrum of clinical entities, including fibroses in multiple organs, hypertrophic scarring following burns, loss of cardiac function following ischemia, and the formation of cancer stroma. However, fibroblasts remain a poorly characterized type of cell, largely due to their inherent heterogeneity. Existing methods for the isolation of fibroblasts require time in cell culture that profoundly influences cell phenotype and behavior. Consequently, many studies investigating fibroblast biology rely upon in vitro manipulation and do not accurately capture fibroblast behavior in vivo. To overcome this problem, we developed a FACS-based protocol for the isolation of fibroblasts from the dorsal skin of adult mice that does not require cell culture, thereby preserving the physiologic transcriptional and proteomic profile of each cell. Our strategy allows for exclusion of non-mesenchymal lineages via a lineage negative gate (Lin(-)) rather than a positive selection strategy to avoid pre-selection or enrichment of a subpopulation of fibroblasts expressing specific surface markers and be as inclusive as possible across this heterogeneous cell type.


Asunto(s)
Fibroblastos/citología , Citometría de Flujo/métodos , Piel/citología , Animales , Matriz Extracelular , Ratones
9.
Tissue Eng Part A ; 22(3-4): 214-21, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26585335

RESUMEN

BACKGROUND: Reconstruction of soft tissue defects has traditionally relied on the use of grafts and flaps, which may be associated with variable resorption and/or significant donor site morbidity. Cell-based strategies employing adipose-derived stromal cells (ASCs), found within the stromal vascular fraction (SVF) of adipose tissue, may offer an alternative strategy for soft tissue reconstruction. In this study, we investigated the potential of a bone morphogenetic protein receptor type 1A (BMPR1A)(+) subpopulation of ASCs to enhance de novo adipogenesis. METHODS: Human lipoaspirate was enzymatically digested to isolate SVF and magnetic-activated cell separation was utilized to obtain BMPR1A(+) and BMPR1A(-) cells. These cells, along with unenriched cells, were expanded in culture and evaluated for adipogenic gene expression and in vitro adipocyte formation. Cells from each group were also labeled with a green fluorescent protein (GFP) lentivirus and transplanted into the inguinal fat pads, an adipogenic niche, of immunocompromised mice to determine their potential for de novo adipogenesis. Confocal microscopy along with staining of lipid droplets and vasculature was performed to evaluate the formation of mature adipocytes by transplanted cells. RESULTS: In comparison to BMPR1A(-) and unenriched ASCs, BMPR1A(+) cells demonstrated significantly enhanced adipogenesis when cultured in an adipogenic differentiation medium, as evidenced by increased staining with Oil Red O and increased expression of peroxisome proliferator-activating receptor gamma (PPAR-γ) and fatty acid-binding protein 4 (FABP4). BMPR1A(+) cells also formed significantly more adipocytes in vivo, as demonstrated by quantification of GFP+ adipocytes. Minimal formation of mature adipocytes was appreciated by BMPR1A(-) cells. CONCLUSIONS: BMPR1A(+) ASCs show an enhanced ability for adipogenesis in vitro, as shown by gene expression and histological staining. Furthermore, within an adipogenic niche, BMPR1A(+) cells possessed an increased capacity to generate de novo fat compared to BMPR1A(-) and unenriched cells. This suggests utility for the BMPR1A(+) subpopulation in cell-based strategies for soft tissue reconstruction.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/biosíntesis , Adipocitos/citología , Tejido Adiposo/citología , Adulto , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Células Cultivadas , Femenino , Humanos , Lentivirus , Ratones , Persona de Mediana Edad , Células del Estroma/citología , Células del Estroma/metabolismo , Transducción Genética
10.
Stem Cells ; 34(3): 668-73, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26661694

RESUMEN

Radiation therapy is not only a mainstay in the treatment of many malignancies but also results in collateral obliteration of microvasculature and dermal/subcutaneous fibrosis. Soft tissue reconstruction of hypovascular, irradiated recipient sites through fat grafting remains challenging; however, a coincident improvement in surrounding skin quality has been noted. Cell-assisted lipotransfer (CAL), the enrichment of fat with additional adipose-derived stem cells (ASCs) from the stromal vascular fraction, has been shown to improve fat volume retention, and enhanced outcomes may also be achieved with CAL at irradiated sites. Supplementing fat grafts with additional ASCs may also augment the regenerative effect on radiation-damaged skin. In this study, we demonstrate the ability for CAL to enhance fat graft volume retention when placed beneath the irradiated scalps of immunocompromised mice. Histologic metrics of fat graft survival were also appreciated, with improved structural qualities and vascularity. Finally, rehabilitation of radiation-induced soft tissue changes were also noted, as enhanced amelioration of dermal thickness, collagen content, skin vascularity, and biomechanical measures were all observed with CAL compared to unsupplemented fat grafts. Supplementation of fat grafts with ASCs therefore shows promise for reconstruction of complex soft tissue defects following adjuvant radiotherapy.


Asunto(s)
Adipocitos/trasplante , Fibrosis/terapia , Células del Estroma/trasplante , Animales , Fibrosis/patología , Supervivencia de Injerto , Humanos , Ratones , Microvasos/patología , Microvasos/efectos de la radiación , Radioterapia/efectos adversos , Piel/patología , Piel/efectos de la radiación
11.
Stem Cells Transl Med ; 5(2): 248-57, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26702129

RESUMEN

Human mesenchymal stem cells (MSCs) have recently become a focus of regenerative medicine, both for their multilineage differentiation capacity and their excretion of proregenerative cytokines. Adipose-derived mesenchymal stem cells (ASCs) are of particular interest because of their abundance in fat tissue and the ease of harvest via liposuction. However, little is known about the impact of different liposuction methods on the functionality of ASCs. Here we evaluate the regenerative abilities of ASCs harvested via a third-generation ultrasound-assisted liposuction (UAL) device versus ASCs obtained via standard suction-assisted lipoaspiration (SAL). Lipoaspirates were sorted using fluorescent assisted cell sorting based on an established surface-marker profile (CD34+/CD31-/CD45-), to obtain viable ASCs. Yield and viability were compared and the differentiation capacities of the ASCs were assessed. Finally, the regenerative potential of ASCs was examined using an in vivo model of tissue regeneration. UAL- and SAL-derived samples demonstrated equivalent ASC yield and viability, and UAL ASCs were not impaired in their osteogenic, adipogenic, or chondrogenic differentiation capacity. Equally, quantitative real-time polymerase chain reaction showed comparable expression of most osteogenic, adipogenic, and key regenerative genes between both ASC groups. Cutaneous regeneration and neovascularization were significantly enhanced in mice treated with ASCs obtained by either UAL or SAL compared with controls, but there were no significant differences in healing between cell-therapy groups. We conclude that UAL is a successful method of obtaining fully functional ASCs for regenerative medicine purposes. Cells harvested with this alternative approach to liposuction are suitable for cell therapy and tissue engineering applications. Significance: Adipose-derived mesenchymal stem cells (ASCs) are an appealing source of therapeutic progenitor cells because of their multipotency, diverse cytokine profile, and ease of harvest via liposuction. Alternative approaches to classical suction-assisted liposuction are gaining popularity; however, little evidence exists regarding the impact of different liposuction methods on the regenerative functionality of ASCs. Human ASC characteristics and regenerative capacity were assessed when harvested via ultrasound-assisted (UAL) versus standard suction-assisted liposuction. ASCs obtained via UAL were of equal quality when directly compared with the current gold standard harvest method. UAL is an adjunctive source of fully functional mesenchymal stem cells for applications in basic research and clinical therapy.


Asunto(s)
Grasa Abdominal/cirugía , Adipocitos/citología , Procedimientos Quirúrgicos Electivos/instrumentación , Lipectomía/instrumentación , Células Madre Mesenquimatosas/citología , Grasa Abdominal/citología , Grasa Abdominal/diagnóstico por imagen , Grasa Abdominal/metabolismo , Adipocitos/metabolismo , Adulto , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Supervivencia Celular , Condrocitos/citología , Condrocitos/metabolismo , Procedimientos Quirúrgicos Electivos/métodos , Femenino , Citometría de Flujo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lipectomía/métodos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Persona de Mediana Edad , Neovascularización Fisiológica , Osteoblastos/citología , Osteoblastos/metabolismo , Ultrasonografía , Cicatrización de Heridas/fisiología
12.
Plast Reconstr Surg ; 136(1): 189-196, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26111321

RESUMEN

BACKGROUND: Plastic surgery is among the most competitive specialties in medicine, but little is known about the attributes of programs that are most attractive to successful applicants. This study aimed to understand and provide insights regarding program characteristics that are most influential to students when ranking plastic surgery programs. METHODS: An anonymous online survey was conducted with newly matched plastic surgery residents for the integrated and combined Match in 2012 and 2013. Subjects were queried regarding their demographics, qualifications, application experiences, and motivations for residency program selection. RESULTS: A total of 92 of 245 matched plastic surgery residents (38 percent) responded to the survey. The perception of resident happiness was the most positive factor influencing program ranking, followed by high operative volume, faculty mentorship, and strong research infrastructure. Perception of a program as "malignant" was the most negative attribute. Applicants with Step 1 scores greater than 245 received significantly more interviews (p =0.001) and considered resident benefits less important (p < 0.05), but geographic location more important (p =0.005). Applicants who published more than two articles also received more interviews (p =0.001) and ranked a strong research infrastructure and program reputation as significantly more important (p < 0.05). Forty-two percent of applicants completed an away rotation at the program with which they matched, and these applicants were more likely to match at their number one ranked program (p = 0.001). CONCLUSIONS: Plastic surgery applicants have differing preferences regarding the ideal training program, but some attributes resonate. These trends can guide programs for improvement in attracting the best applicants.


Asunto(s)
Actitud del Personal de Salud , Selección de Profesión , Educación de Postgrado en Medicina , Internado y Residencia , Cirugía Plástica/educación , Estudios Transversales , Recolección de Datos , Femenino , Humanos , Masculino , Mentores , Motivación , Estados Unidos
13.
Plast Reconstr Surg ; 136(4): 752-763, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26090763

RESUMEN

BACKGROUND: Adipose-derived stromal cells represent a relatively abundant source of multipotent cells, with many potential applications in regenerative medicine. The present study sought to demonstrate the use of RNA sequencing in identifying differentially expressed transcripts, particularly long noncoding RNAs, associated with adipogenic differentiation to gain a clearer picture of the mechanisms responsible for directing adipose-derived stromal cell fate toward the adipogenic lineage. METHODS: Human adipose-derived stromal cells were cultured in adipogenic differentiation media, and RNA was harvested at days 0, 1, 3, 5, and 7. Directional RNA sequencing libraries were prepared and sequenced. Paired-end reads were mapped to the human genome reference sequence hg19. Transcriptome assembly was performed and significantly differentially expressed transcripts were identified. Gene ontology term analysis was then performed to identify coding and noncoding transcripts of interest. Differential expression was verified by quantitative real-time polymerase chain reaction. RESULTS: Of 2868 significantly differentially expressed transcripts identified, 207 were noncoding. Enriched gene ontology terms among up-regulated coding transcripts notably reflected differentiation toward the adipogenic lineage. Enriched gene ontology terms among down-regulated coding transcripts reflected growth arrest. Guilt-by-association analysis revealed noncoding RNA candidates with potential roles in the process of adipogenic differentiation. CONCLUSIONS: The precise mechanisms that guide lineage-specific differentiation in multipotent cells are not yet fully understood. Defining long noncoding RNAs associated with adipogenic differentiation allows for potential manipulation of regulatory pathways in novel ways. The authors present RNA sequencing as a powerful tool for expanding the understanding of adipose-derived stromal cells and developing novel applications within regenerative medicine.


Asunto(s)
Adipocitos/fisiología , Adipogénesis/genética , Perfilación de la Expresión Génica , ARN Largo no Codificante , Análisis de Secuencia de ARN , Células del Estroma/fisiología , Transcriptoma , Adulto , Femenino , Humanos , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Tissue Eng Part A ; 21(15-16): 2281-91, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26020286

RESUMEN

Adipose tissue contains an abundant source of multipotent mesenchymal cells termed "adipose-derived stromal cells" (ASCs) that hold potential for regenerative medicine. However, the heterogeneity inherent to ASCs harvested using standard methodologies remains largely undefined, particularly in regards to differences across donors. Identifying the subpopulations of ASCs predisposed toward differentiation along distinct lineages holds value for improving graft survival, predictability, and efficiency. Human ASCs (hASCs) from three different donors were independently isolated by density-based centrifugation from adipose tissue and maintained in culture or differentiated along either adipogenic or osteogenic lineages using differentiation media. Undifferentiated and differentiated hASCs were then analyzed for the presence of 242 human surface markers by flow cytometry analysis. By comprehensively characterizing the surface marker profile of undifferentiated hASCs using flow cytometry, we gained novel insights into the heterogeneity underlying protein expression on the surface of cultured undifferentiated hASCs across different donors. Comparison of the surface marker profile of undifferentiated hASCs with hASCs that have undergone osteogenic or adipogenic differentiation allowed for the identification of surface markers that were upregulated and downregulated by osteogenic or adipogenic differentiation. Osteogenic differentiation induced upregulation of CD164 and downregulation of CD49a, CD49b, CD49c, CD49d, CD55, CD58, CD105, and CD166 while adipogenic differentiation induced upregulation of CD36, CD40, CD146, CD164, and CD271 and downregulation of CD49b, CD49c, CD49d, CD71, CD105, and CD166. These results lend support to the notion that hASCs isolated using standard methodologies represent a heterogeneous population and serve as a foundation for future studies seeking to maximize their regenerative potential through fluorescence-activated cell sorting-based selection before therapy.


Asunto(s)
Tejido Adiposo/metabolismo , Antígenos de Diferenciación/biosíntesis , Diferenciación Celular , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Tejido Adiposo/citología , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología
15.
Plast Reconstr Surg ; 136(1): 67-75, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25829158

RESUMEN

BACKGROUND: Cell-assisted lipotransfer has shown much promise as a technique for improving fat graft take. However, the concentration of stromal vascular fraction cells required to optimally enhance fat graft retention remains unknown. METHODS: Human lipoaspirate was processed for both fat transfer and harvest of stromal vascular fraction cells. Cells were then mixed back with fat at varying concentrations ranging from 10,000 to 10 million cells per 200 µl of fat. Fat graft volume retention was assessed by means of computed tomographic scanning over 8 weeks, and then fat grafts were explanted and compared histologically for overall architecture and vascularity. RESULTS: Maximum fat graft retention was seen at a concentration of 10,000 cells per 200 µl of fat. The addition of higher number of cells negatively impacted fat graft retention, with supplementation of 10 million cells producing the lowest final volumes, lower than fat alone. Interestingly, fat grafts supplemented with 10,000 cells showed significantly increased vascularity and decreased inflammation, whereas fat grafts supplemented with 10 million cells showed significant lipodegeneration compared with fat alone CONCLUSIONS: : The authors' study demonstrates dose dependence in the number of stromal vascular fraction cells that can be added to a fat graft to enhance retention. Although cell-assisted lipotransfer may help promote graft survival, this effect may need to be balanced with the increased metabolic load of added cells that may compete with adipocytes for nutrients during the postgraft period.


Asunto(s)
Adipocitos/trasplante , Supervivencia de Injerto , Grasa Subcutánea/trasplante , Adipocitos/patología , Adulto , Animales , Femenino , Humanos , Ratones , Células del Estroma/trasplante , Grasa Subcutánea/patología
16.
Nanomedicine ; 11(5): 1253-63, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25791811

RESUMEN

Nanotechnology represents a major frontier with potential to significantly advance the field of bone tissue engineering. Current limitations in regenerative strategies include impaired cellular proliferation and differentiation, insufficient mechanical strength of scaffolds, and inadequate production of extrinsic factors necessary for efficient osteogenesis. Here we review several major areas of research in nanotechnology with potential implications in bone regeneration: 1) nanoparticle-based methods for delivery of bioactive molecules, growth factors, and genetic material, 2) nanoparticle-mediated cell labeling and targeting, and 3) nano-based scaffold construction and modification to enhance physicochemical interactions, biocompatibility, mechanical stability, and cellular attachment/survival. As these technologies continue to evolve, ultimate translation to the clinical environment may allow for improved therapeutic outcomes in patients with large bone deficits and osteodegenerative diseases. FROM THE CLINICAL EDITOR: Traditionally, the reconstruction of bony defects has relied on the use of bone grafts. With advances in nanotechnology, there has been significant development of synthetic biomaterials. In this article, the authors provided a comprehensive review on current research in nanoparticle-based therapies for bone tissue engineering, which should be useful reading for clinicians as well as researchers in this field.


Asunto(s)
Regeneración Ósea , Nanotecnología/métodos , Ingeniería de Tejidos/métodos , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Humanos , Nanomedicina/métodos , Nanopartículas/química , Nanopartículas/metabolismo , Medicina Regenerativa/métodos , Coloración y Etiquetado/métodos , Andamios del Tejido/química
17.
Expert Opin Emerg Drugs ; 20(2): 235-46, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25704608

RESUMEN

INTRODUCTION: Wound healing can be characterized as underhealing, as in the setting of chronic wounds, or overhealing, occurring with hypertrophic scar formation after burn injury. Topical therapies targeting specific biochemical and molecular pathways represent a promising avenue for improving and, in some cases normalizing, the healing process. AREAS COVERED: A brief overview of both normal and pathological wound healing has been provided, along with a review of the current clinical guidelines and treatment modalities for chronic wounds, burn wounds and scar formation. Next, the major avenues for wound healing drugs, along with drugs currently in development, are discussed. Finally, potential challenges to further drug development, and future research directions are discussed. EXPERT OPINION: The large body of research concerning wound healing pathophysiology has provided multiple targets for topical therapies. Growth factor therapies with the ability to be targeted for localized release in the wound microenvironment are most promising, particularly when they modulate processes in the proliferative phase of wound healing.


Asunto(s)
Diseño de Fármacos , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/tratamiento farmacológico , Administración Tópica , Animales , Quemaduras/complicaciones , Quemaduras/tratamiento farmacológico , Quemaduras/patología , Cicatriz/tratamiento farmacológico , Cicatriz/etiología , Humanos , Terapia Molecular Dirigida , Guías de Práctica Clínica como Asunto , Heridas y Lesiones/patología
18.
J Vis Exp ; (95): 52181, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25650785

RESUMEN

Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are considered the gold standard for stem cell-based tissue engineering applications. However, the process by which they must be harvested can be associated with significant donor site morbidity. In contrast, adipose-derived stromal cells (ASCs) are more readily abundant and more easily harvested, making them an appealing alternative to BM-MSCs. Like BM-MSCs, ASCs can differentiate into osteogenic lineage cells and can be used in tissue engineering applications, such as seeding onto scaffolds for use in craniofacial skeletal defects. ASCs are obtained from the stromal vascular fraction (SVF) of digested adipose tissue, which is a heterogeneous mixture of ASCs, vascular endothelial and mural cells, smooth muscle cells, pericytes, fibroblasts, and circulating cells. Flow cytometric analysis has shown that the surface marker profile for ASCs is similar to that for BM-MSCs. Despite several published reports establishing markers for the ASC phenotype, there is still a lack of consensus over profiles identifying osteoprogenitor cells in this heterogeneous population. This protocol describes how to isolate and use a subpopulation of ASCs with enhanced osteogenic capacity to repair critical-sized calvarial defects.


Asunto(s)
Tejido Adiposo/citología , Citometría de Flujo/métodos , Células Madre Mesenquimatosas/citología , Osteogénesis/fisiología , Adipocitos/citología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Linaje de la Célula , Femenino , Humanos , Fenotipo
19.
Plast Reconstr Surg ; 135(3): 907-917, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25719706

RESUMEN

Over 100 million patients acquire scars in the industrialized world each year, primarily as a result of elective operations. Although undefined, the global incidence of scarring is even larger, extending to significant numbers of burn and other trauma-related wounds. Scars have the potential to exert a profound psychological and physical impact on the individual. Beyond aesthetic considerations and potential disfigurement, scarring can result in restriction of movement and reduced quality of life. The formation of a scar following skin injury is a consequence of wound healing occurring through reparative rather than regenerative mechanisms. In this article, the authors review the basic stages of wound healing; differences between adult and fetal wound healing; various mechanical, genetic, and pharmacologic strategies to reduce scarring; and the biology of skin stem/progenitor cells that may hold the key to scarless regeneration.


Asunto(s)
Cicatriz/fisiopatología , Regeneración/fisiología , Fenómenos Fisiológicos de la Piel , Piel/fisiopatología , Cicatrización de Heridas/fisiología , Humanos
20.
J Vis Exp ; (95): e52217, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25590561

RESUMEN

Lipotransfer is a vital tool in the surgeon's armamentarium for the treatment of soft tissue deficits of throughout the body. Fat is the ideal soft tissue filler as it is readily available, easily obtained, inexpensive, and inherently biocompatible.(1) However, despite its burgeoning popularity, fat grafting is hampered by unpredictable results and variable graft survival, with published retention rates ranging anywhere from 10-80%. (1-3) To facilitate investigations on fat grafting, we have therefore developed an animal model that allows for real-time analysis of injected fat volume retention. Briefly, a small cut is made in the scalp of a CD-1 nude mouse and 200-400 µl of processed lipoaspirate is placed over the skull. The scalp is chosen as the recipient site because of its absence of native subcutaneous fat, and because of the excellent background contrast provided by the calvarium, which aids in the analysis process. Micro-computed tomography (micro-CT) is used to scan the graft at baseline and every two weeks thereafter. The CT images are reconstructed, and an imaging software is used to quantify graft volumes. Traditionally, techniques to assess fat graft volume have necessitated euthanizing the study animal to provide just a single assessment of graft weight and volume by physical measurement ex vivo. Biochemical and histological comparisons have likewise required the study animal to be euthanized. This described imaging technique offers the advantage of visualizing and objectively quantifying volume at multiple time points after initial grafting without having to sacrifice the study animal. The technique is limited by the size of the graft able to be injected as larger grafts risk skin and fat necrosis. This method has utility for all studies evaluating fat graft viability and volume retention. It is particularly well-suited to providing a visual representation of fat grafts and following changes in volume over time.


Asunto(s)
Tejido Adiposo/trasplante , Supervivencia de Injerto/fisiología , Trasplante Heterólogo/métodos , Animales , Femenino , Humanos , Ratones , Ratones Desnudos , Modelos Animales , Microtomografía por Rayos X/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA