Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Exp Dermatol ; 28(4): 485-492, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-28677217

RESUMEN

Reactive oxygen species (ROS) impair wound healing through destructive oxidation of intracellular proteins, lipids and nucleic acids. Intracellular superoxide dismutase (SOD1) regulates ROS levels and plays a critical role in tissue homoeostasis. Recent evidence suggests that age-associated wound healing impairments may partially result from decreased SOD1 expression. We investigated the mechanistic basis by which increased oxidative stress links to age-associated impaired wound healing. Fibroblasts were isolated from unwounded skin of young and aged mice, and myofibroblast differentiation was assessed by measuring α-smooth muscle actin and collagen gel contraction. Excisional wounds were created on young and aged mice to study the healing rate, ROS levels and SOD1 expression. A mechanistic link between oxidative stress and fibroblast function was explored by assessing the TGF-ß1 signalling pathway components in young and aged mice. Age-related wounds displayed reduced myofibroblast differentiation and delayed wound healing, consistent with a decrease in the in vitro capacity for fibroblast-myofibroblast transition following oxidative stress. Young fibroblasts with normal SOD1 expression exhibited increased phosphorylation of ERK in response to elevated ROS. In contrast, aged fibroblasts with reduced SOD1 expression displayed a reduced capacity to modulate intracellular ROS. Collectively, age-associated wound healing impairments are associated with fibroblast dysfunction that is likely the result of decreased SOD1 expression and subsequent dysregulation of intracellular ROS. Strategies targeting these mechanisms may suggest a new therapeutic approach in the treatment of chronic non-healing wounds in the aged population.


Asunto(s)
Envejecimiento/metabolismo , Fibroblastos/fisiología , Superóxido Dismutasa-1/deficiencia , Cicatrización de Heridas , Animales , Diferenciación Celular , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos C57BL , Estrés Oxidativo
2.
Exp Dermatol ; 25(3): 206-11, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26663425

RESUMEN

Advanced age is characterized by impairments in wound healing, and evidence is accumulating that this may be due in part to a concomitant increase in oxidative stress. Extended exposure to reactive oxygen species (ROS) is thought to lead to cellular dysfunction and organismal death via the destructive oxidation of intra-cellular proteins, lipids and nucleic acids. Extracellular superoxide dismutase (ecSOD/SOD3) is a prime antioxidant enzyme in the extracellular space that eliminates ROS. Here, we demonstrate that reduced SOD3 levels contribute to healing impairments in aged mice. These impairments include delayed wound closure, reduced neovascularization, impaired fibroblast proliferation and increased neutrophil recruitment. We further establish that SOD3 KO and aged fibroblasts both display reduced production of TGF-ß1, leading to decreased differentiation of fibroblasts into myofibroblasts. Taken together, these results suggest that wound healing impairments in ageing are associated with increased levels of ROS, decreased SOD3 expression and impaired extracellular oxidative stress regulation. Our results identify SOD3 as a possible target to correct age-related cellular dysfunction in wound healing.


Asunto(s)
Envejecimiento , Fibroblastos/efectos de los fármacos , Neovascularización Fisiológica , Superóxido Dismutasa/deficiencia , Cicatrización de Heridas , Animales , Antioxidantes/metabolismo , Proliferación Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neutrófilos/citología , Estrés Oxidativo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
3.
Ann Surg ; 260(6): 1138-46, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25389925

RESUMEN

OBJECTIVE: To investigate how epithelial mechanotransduction pathways impact wound repair. BACKGROUND: Mechanical forces are increasingly recognized to influence tissue repair, but their role in chronic wound pathophysiology remains unknown. Studies have shown that chronic wounds exhibit high levels of matrix metalloproteinase 9 (MMP9), a key proteolytic enzyme that regulates wound remodeling. We hypothesized that epithelial mechanosensory pathways regulated by keratinocyte-specific focal adhesion kinase (FAK) control dermal remodeling via MMP9. METHODS: A standard wound model was applied to keratinocyte-specific FAK knockout (KO) and control mice. Rates of wound healing were measured and tissue was obtained for histologic and molecular analyses. Transcriptional and immunoblot assays were used to assess the activation of FAK, intracellular kinases, and MMP9 in vitro. A cell suspension model was designed to validate the importance of FAK mechanosensing, p38, and MMP9 secretion in human cells. Biomechanical testing was utilized to evaluate matrix tensile properties in FAK KO and control wounds. RESULTS: Wound healing in FAK KO mice was significantly delayed compared with controls (closure at 15 days compared with 20 days, P = 0.0003). FAK KO wounds demonstrated decreased dermal thickness and collagen density. FAK KO keratinocytes exhibited overactive p38 and MMP9 signaling in vitro, findings recapitulated in human keratinocytes via the deactivation of FAK in the cell suspension model. Functionally, FAK KO wounds were significantly weaker and more brittle than control wounds, results consistent with the histologic and molecular analyses. CONCLUSIONS: Keratinocyte FAK is highly responsive to mechanical cues and may play a critical role in matrix remodeling via regulation of p38 and MMP9. These findings suggest that aberrant epithelial mechanosensory pathways may contribute to pathologic dermal proteolysis and wound chronicity.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/genética , Queratinocitos/ultraestructura , ARN/genética , Piel/lesiones , Regulación hacia Arriba , Cicatrización de Heridas , Heridas y Lesiones/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Proteína-Tirosina Quinasas de Adhesión Focal/biosíntesis , Humanos , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Recién Nacido , Queratinocitos/metabolismo , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Proteolisis , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología
4.
Differentiation ; 86(3): 87-91, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23623400

RESUMEN

Historically, great efforts have been made to elucidate the biochemical pathways that direct the complex process of wound healing; however only recently has there been recognition of the importance that mechanical signals play in the process of tissue repair and scar formation. The body's physiologic response to injury involves a dynamic interplay between mechanical forces and biochemical cues which directs a cascade of signals leading ultimately to the formation of fibrotic scar. Fibroblasts are a highly mechanosensitive cell type and are also largely responsible for the generation of the fibrotic matrix during scar formation and are thus a critical player in the process of mechanotransduction during tissue repair. Mechanotransduction is initiated at the interface between the cell membrane and the extracellular matrix where mechanical signals are first translated into a biochemical response. Focal adhesions are dynamic multi-protein complexes through which the extracellular matrix links to the intracellular cytoskeleton. These focal adhesion complexes play an integral role in the propagation of this initial mechanical cue into an extensive network of biochemical signals leading to widespread downstream effects including the influx of inflammatory cells, stimulation of angiogenesis, keratinocyte migration, fibroblast proliferation and collagen synthesis. Increasing evidence has demonstrated the importance of the biomechanical milieu in healing wounds and suggests that an integrated approach to the discovery of targets to decrease scar formation may prove more clinically efficacious than previous purely biochemical strategies.


Asunto(s)
Cicatriz/metabolismo , Fibroblastos/metabolismo , Adhesiones Focales/metabolismo , Mecanotransducción Celular , Animales , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos
5.
FASEB J ; 25(12): 4498-510, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21911593

RESUMEN

Mechanical force significantly modulates both inflammation and fibrosis, yet the fundamental mechanisms that regulate these interactions remain poorly understood. Here we performed microarray analysis to compare gene expression in mechanically loaded wounds vs. unloaded control wounds in an established murine hypertrophic scar (HTS) model. We identified 853 mechanically regulated genes (false discovery rate <2) at d 14 postinjury, a subset of which were enriched for T-cell-regulated pathways. To substantiate the role of T cells in scar mechanotransduction, we applied the HTS model to T-cell-deficient mice and wild-type mice. We found that scar formation in T-cell-deficient mice was reduced by almost 9-fold (P < 0.001) with attenuated epidermal (by 2.6-fold, P < 0.01) and dermal (3.9-fold, P < 0.05) proliferation. Mechanical stimulation was highly associated with sustained T-cell-dependent Th2 cytokine (IL-4 and IL-13) and chemokine (MCP-1) signaling. Further, T-cell-deficient mice failed to recruit systemic inflammatory cells such as macrophages or monocytic fibroblast precursors in response to mechanical loading. These findings indicate that T-cell-regulated fibrogenic pathways are highly mechanoresponsive and suggest that mechanical forces induce a chronic-like inflammatory state through immune-dependent activation of both local and systemic cell populations.


Asunto(s)
Cicatriz Hipertrófica/complicaciones , Cicatriz Hipertrófica/inmunología , Inflamación/etiología , Inflamación/inmunología , Linfocitos T/inmunología , Animales , Proliferación Celular , Cicatriz Hipertrófica/genética , Cicatriz Hipertrófica/patología , Citocinas/genética , Citocinas/metabolismo , Femenino , Fibrosis , Expresión Génica , Inflamación/genética , Inflamación/patología , Activación de Macrófagos , Mecanotransducción Celular/genética , Mecanotransducción Celular/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Transducción de Señal , Estrés Mecánico , Cicatrización de Heridas/genética , Cicatrización de Heridas/inmunología
6.
Wound Repair Regen ; 19(1): 49-58, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21134033

RESUMEN

Although numerous factors are implicated in skin fibrosis, the exact pathophysiology of hypertrophic scarring remains unknown. We recently demonstrated that mechanical force initiates hypertrophic scar formation in a murine model, potentially enhancing cellular survival through Akt. Here, we specifically examined Akt-mediated mechanotransduction in fibroblasts using both strain culture systems and our murine scar model. In vitro, static strain increased fibroblast motility, an effect blocked by wortmannin (a phosphoinositide-3-kinase/Akt inhibitor). We also demonstrated that high-frequency cyclic strain was more effective at inducing Akt phosphorylation than low frequency or static strain. In vivo, Akt phosphorylation was induced by mechanical loading of dermal fibroblasts in both unwounded and wounded murine skin. Mechanically loaded scars also exhibited strong expression of α-smooth muscle actin, a putative marker of pathologic scar formation. In vivo inhibition of Akt increased apoptosis but did not significantly abrogate hypertrophic scar development. These data suggest that although Akt signaling is activated in fibroblasts during mechanical loading of skin, this is not the critical pathway in hypertrophic scar formation. Future studies are needed to fully elucidate the critical mechanotransduction components and pathways which activate skin fibrosis.


Asunto(s)
Cicatriz Hipertrófica/enzimología , Cicatriz Hipertrófica/etiología , Fibroblastos/fisiología , Mecanotransducción Celular/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Androstadienos/farmacología , Animales , Movimiento Celular , Supervivencia Celular , Cicatriz Hipertrófica/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Estrés Mecánico , Wortmanina
7.
J Plast Reconstr Aesthet Surg ; 72(12): 2056-2063, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31648962

RESUMEN

INTRODUCTION: Bone grafting of alveolar clefts is routinely performed with cancellous bone harvested from the iliac crest. Graft site morbidity is frequently seen, with early postoperative pain being one of the most common complaints. Liposomal bupivacaine (LB) has been demonstrated to provide improvement in postoperative pain for patients undergoing bunionectomy or hemorrhoidectomy, which may translate to patients requiring iliac crest bone graft harvest. METHODS: Thirty-eight patients undergoing iliac crest bone harvest were included in the study. Twenty-one patients underwent open iliac crest bone graft harvest with administration of 0.25% bupivacaine at the hip donor site, while 17 patients received local infiltration of 1.3% liposomal bupivacaine. Patient-reported pain scores, total narcotic use, length of stay, and postoperative steps were monitored. RESULTS: There were no significant differences in age, weight, distribution of clefts, or choice of donor hip between the two groups. There were no significant differences in length of hospitalization stay. However, differences were noted in average postoperative pain scores at five of six time points in the first 24 h, total oral morphine equivalents administered (4.7 ±â€¯5.3 vs. 14.3 ±â€¯12.0), and steps at postoperative days one to three (p < 0.001, for all three days) for patients receiving 1.3% LB versus 0.25% bupivacaine, respectively. CONCLUSION: Reduced pain scores and increased postoperative activity highlight the potential of LB to improve postoperative pain management in children undergoing iliac crest bone harvest for alveolar bone grafting.


Asunto(s)
Injerto de Hueso Alveolar/métodos , Anestésicos Locales/administración & dosificación , Bupivacaína/administración & dosificación , Labio Leporino/cirugía , Fisura del Paladar/cirugía , Dolor Postoperatorio/prevención & control , Administración Tópica , Analgesia/métodos , Niño , Femenino , Humanos , Ilion/trasplante , Cuidados Intraoperatorios/métodos , Tiempo de Internación/estadística & datos numéricos , Liposomas , Medición de Resultados Informados por el Paciente , Estudios Retrospectivos , Sitio Donante de Trasplante
8.
Burns ; 44(5): 1203-1209, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29728283

RESUMEN

INTRODUCTION: Feeding tubes in burn patients are at high risk for becoming dislodged as traditional tape securement does not adhere well to sloughed skin, resulting in nutrition delivery disruption and placing patients at increased risk for iatrogenic injury upon reinsertion. METHODS: Seventy-four patients admitted to our regional burn center requiring nasoenteric nutritional support were prospectively followed. Fourty-one patients received a nasal bridle while thirty-three received traditional tape and elastic dressings. Primary outcomes centered on measuring clinical efficacy of the nasal bridle system. RESULTS: Conventional tape-secured feeding tubes were dislodged more frequently (0.9±0.2 times per 10 feeding days vs. 0.2±0.1 times per 10 feeding days; p=0.005). Nasal bridle secured tubes showed significantly longer functional life on Kaplan Meier analysis (hazard ratio 0.35; p=0.01). Fewer abdominal x-ray studies were performed to confirm tube placement in nasal bridle patients (1.48±0.13 for nasal bridle vs. 2.21±0.21 for conventional tape-secured; p=0.003). Overall, patients with bridle securement had fewer hours of missed enteric feeds (2.51±0.95hours vs. 6.72±2.07hours; p=0.05). Importantly, utilization of a nasal bridle decreased overall estimated costs for enteric feeding management ($1,379.72±120.70 vs. $1,107.66±63.95; p=0.05). CONCLUSIONS: Utilization of a nasal bridle system provides a reliable method for securement of nasoenteric feeding tubes with clinical benefits in the burn patient population.


Asunto(s)
Quemaduras/terapia , Nutrición Enteral/instrumentación , Falla de Equipo/estadística & datos numéricos , Intubación Gastrointestinal/instrumentación , Adulto , Nutrición Enteral/métodos , Femenino , Humanos , Incidencia , Intubación Gastrointestinal/métodos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Estudios Prospectivos
9.
Plast Reconstr Surg ; 138(3 Suppl): 31S-41S, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27556772

RESUMEN

BACKGROUND: Chronic wounds present unique challenges for healthcare providers as they place patients at increased risk for various morbidities and mortality. Advances in wound care technology have expanded the treatment options available for wound management, but few products fully address the underlying core deficiencies responsible for the development of poorly healing wounds. In the future, addressing these derangements will undoubtedly play a key role in the treatment of these patients. Broad enthusiasm has surrounded the field of stem cell biology, which has shown great promise in repairing damaged tissues across numerous disease phenotypes. METHODS: In this review, we provide a comprehensive review of the literature and evaluate the present landscape of wound therapeutics while discussing the rationales and allure behind stem cell-based products. We further propose 2 challenges that remain as new stem cell-based therapies are being developed and as this technology moves toward clinical translation. RESULTS: Given the relatively young age of this newer technology in wound healing, numerous challenges continue to surround its effective use including identifying the ideal population of stem cells to use and determining the optimal cell delivery method. However, significant forward progress has been made, with several clinical trials beginning to demonstrate reliable clinical benefit. CONCLUSION: The upward trajectory of stem cell technologies provides an exciting opportunity to positively impact patient outcomes through the controlled application of regenerative cell-based therapy.


Asunto(s)
Trasplante de Células Madre/métodos , Herida Quirúrgica/terapia , Cicatrización de Heridas , Humanos , Herida Quirúrgica/fisiopatología , Cicatrización de Heridas/fisiología
10.
Adv Wound Care (New Rochelle) ; 1(4): 147-152, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24527296

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) have shown significant therapeutic potential in preclinical animal models of wound healing. However, the translation of MSC-based therapeutics to clinical practice has been delayed by questions including the mechanisms of MSC homing, engraftment, and ultimate function. THE PROBLEM: Experimental models of MSC-based wound therapies often involve intravenous injection of cells followed by sacrifice of animals at various time points and detection of MSCs in wounds by histological methods. However, this methodology is limited by its sampling of only specific tissue at a single time point and provides no information about how exogenously transplanted MSCs home to the wound environment. BASIC/CLINICAL SCIENCE ADVANCES: Most systemically injected MSCs initially become entrapped within the lungs before migrating out to the liver and spleen in the normal state. When an injury is present, after the initial lung entrapment, MSCs migrate in response to inflammatory mediators and home to sites of wounding. CLINICAL CARE RELEVANCE: As MSC-based wound therapies continue to advance toward clinical trials, the availability of noninvasive methods to track cells after injection into patients affords the opportunity to monitor stem cell behavior post-transplantation. CONCLUSION: MSCs have demonstrated great promise as an emerging therapeutic for wound management. However, further preclinical studies will be needed to elucidate the reparative mechanisms of these cells and to determine how to optimize their regenerative potential.

11.
Biomaterials ; 33(1): 80-90, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21963148

RESUMEN

In this study, we examined the capacity of a biomimetic pullulan-collagen hydrogel to create a functional biomaterial-based stem cell niche for the delivery of mesenchymal stem cells (MSCs) into wounds. Murine bone marrow-derived MSCs were seeded into hydrogels and compared to MSCs grown in standard culture conditions. Hydrogels induced MSC secretion of angiogenic cytokines and expression of transcription factors associated with maintenance of pluripotency and self-renewal (Oct4, Sox2, Klf4) when compared to MSCs grown in standard conditions. An excisonal wound healing model was used to compare the ability of MSC-hydrogel constructs versus MSC injection alone to accelerate wound healing. Injection of MSCs did not significantly improve time to wound closure. In contrast, wounds treated with MSC-seeded hydrogels showed significantly accelerated healing and a return of skin appendages. Bioluminescence imaging and FACS analysis of luciferase+/GFP+ MSCs indicated that stem cells delivered within the hydrogel remained viable longer and demonstrated enhanced engraftment efficiency than those delivered via injection. Engrafted MSCs were found to differentiate into fibroblasts, pericytes and endothelial cells but did not contribute to the epidermis. Wounds treated with MSC-seeded hydrogels demonstrated significantly enhanced angiogenesis, which was associated with increased levels of VEGF and other angiogenic cytokines within the wounds. Our data suggest that biomimetic hydrogels provide a functional niche capable of augmenting MSC regenerative potential and enhancing wound healing.


Asunto(s)
Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Células Madre Mesenquimatosas/citología , Animales , Biomimética , Western Blotting , Diferenciación Celular/fisiología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibroblastos/citología , Citometría de Flujo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Cicatrización de Heridas/efectos de los fármacos
12.
Macromol Biosci ; 11(11): 1458-66, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21994074

RESUMEN

Cell-based therapies for wound repair are limited by inefficient delivery systems that fail to protect cells from the acute inflammatory environment. Here, a biomimetic hydrogel system is described that is based on the polymer pullulan, a carbohydrate glucan known to exhibit potent antioxidant capabilities. It is shown that pullulan hydrogels are an effective cell delivery system and improve mesenchymal stem cell survival and engraftment in high-oxidative-stress environments. The results suggest that glucan hydrogel systems may prove beneficial for progenitor-cell-based approaches to skin regeneration.


Asunto(s)
Antioxidantes/metabolismo , Biomimética/métodos , Glucanos/metabolismo , Hidrogeles/metabolismo , Traumatismos de los Tejidos Blandos , Cicatrización de Heridas/fisiología , Animales , Antioxidantes/síntesis química , Antioxidantes/farmacología , Materiales Biocompatibles/síntesis química , Materiales Biocompatibles/metabolismo , Materiales Biocompatibles/farmacología , Femenino , Glucanos/síntesis química , Glucanos/farmacología , Hidrogeles/síntesis química , Peróxido de Hidrógeno/efectos adversos , Peróxido de Hidrógeno/farmacología , Espectroscopía de Resonancia Magnética , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Regeneración/efectos de los fármacos , Traumatismos de los Tejidos Blandos/metabolismo , Traumatismos de los Tejidos Blandos/terapia , Cicatrización de Heridas/efectos de los fármacos
13.
Tissue Eng Part A ; 17(5-6): 631-44, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20919949

RESUMEN

New strategies for skin regeneration are needed to address the significant medical burden caused by cutaneous wounds and disease. In this study, pullulan-collagen composite hydrogel matrices were fabricated using a salt-induced phase inversion technique, resulting in a structured yet soft scaffold for skin engineering. Salt crystallization induced interconnected pore formation, and modification of collagen concentration permitted regulation of scaffold pore size. Hydrogel architecture recapitulated the reticular distribution of human dermal matrix while maintaining flexible properties essential for skin applications. In vitro, collagen hydrogel scaffolds retained their open porous architecture and viably sustained human fibroblasts and murine mesenchymal stem cells and endothelial cells. In vivo, hydrogel-treated murine excisional wounds demonstrated improved wound closure, which was associated with increased recruitment of stromal cells and formation of vascularized granulation tissue. In conclusion, salt-induced phase inversion techniques can be used to create modifiable pullulan-collagen composite dermal scaffolds that augment early wound healing. These novel biomatrices can potentially serve as a structured delivery template for cells and biomolecules in regenerative skin applications.


Asunto(s)
Colágeno/farmacología , Dermis/efectos de los fármacos , Glucanos/farmacología , Hidrogeles/farmacología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Reactivos de Enlaces Cruzados/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neovascularización Fisiológica/efectos de los fármacos , Porosidad/efectos de los fármacos , Implantación de Prótesis , Ratas , Tejido Subcutáneo/efectos de los fármacos
14.
Nat Med ; 18(1): 148-52, 2011 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-22157678

RESUMEN

Exuberant fibroproliferation is a common complication after injury for reasons that are not well understood. One key component of wound repair that is often overlooked is mechanical force, which regulates cell-matrix interactions through intracellular focal adhesion components, including focal adhesion kinase (FAK). Here we report that FAK is activated after cutaneous injury and that this process is potentiated by mechanical loading. Fibroblast-specific FAK knockout mice have substantially less inflammation and fibrosis than control mice in a model of hypertrophic scar formation. We show that FAK acts through extracellular-related kinase (ERK) to mechanically trigger the secretion of monocyte chemoattractant protein-1 (MCP-1, also known as CCL2), a potent chemokine that is linked to human fibrotic disorders. Similarly, MCP-1 knockout mice form minimal scars, indicating that inflammatory chemokine pathways are a major mechanism by which FAK mechanotransduction induces fibrosis. Small-molecule inhibition of FAK blocks these effects in human cells and reduces scar formation in vivo through attenuated MCP-1 signaling and inflammatory cell recruitment. These findings collectively indicate that physical force regulates fibrosis through inflammatory FAK-ERK-MCP-1 pathways and that molecular strategies targeting FAK can effectively uncouple mechanical force from pathologic scar formation.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Mecanotransducción Celular , Piel/enzimología , Piel/patología , Animales , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Cicatriz Hipertrófica/enzimología , Cicatriz Hipertrófica/patología , Fibroblastos/metabolismo , Fibrosis , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal
15.
Plast Reconstr Surg ; 126(3): 858-868, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20811219

RESUMEN

SUMMARY: Novel tissue- and organ-engineering strategies are needed to address the growing need for replacement biological parts. Collective progress in stem cell technology, biomaterials, engineering, and molecular medicine has advanced the state of regenerative medicine, yet many hurdles to clinical translation remain. Plastic surgeons are in an ideal position to capitalize on emerging technologies and will be at the forefront of transitioning basic science research into the clinical reconstructive arena. This review highlights fundamental principles of bioengineering, recent progress in tissue-specific engineering, and future directions for this exciting and rapidly evolving area of medicine.


Asunto(s)
Procedimientos de Cirugía Plástica/métodos , Ingeniería de Tejidos/métodos , Animales , Humanos
16.
Organogenesis ; 6(3): 151-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21197216

RESUMEN

The field of tissue engineering has made considerable strides since it was first described in the late 1980s. The advent and subsequent boom in stem cell biology, emergence of novel technologies for biomaterial development and further understanding of developmental biology have contributed to this accelerated progress. However, continued efforts to translate tissue-engineering strategies into clinical therapies have been hampered by the problems associated with scaling up laboratory methods to produce large, complex tissues. The significant challenges faced by tissue engineers include the production of an intact vasculature within a tissue-engineered construct and recapitulation of the size and complexity of a whole organ. Here we review the basic components necessary for bioengineering organs-biomaterials, cells and bioactive molecules-and discuss various approaches for augmenting these principles to achieve organ level tissue engineering. Ultimately, the successful translation of tissue-engineered constructs into everyday clinical practice will depend upon the ability of the tissue engineer to "scale up" every aspect of the research and development process.


Asunto(s)
Técnicas de Cultivo de Órganos , Ingeniería de Tejidos , Andamios del Tejido , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA