Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 106(32): 13505-10, 2009 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-19666581

RESUMEN

Diabetes is associated with poor outcomes following acute vascular occlusive events. This results in part from a failure to form adequate compensatory microvasculature in response to ischemia. Since vascular endothelial growth factor (VEGF) is an essential mediator of neovascularization, we examined whether hypoxic up-regulation of VEGF was impaired in diabetes. Both fibroblasts isolated from type 2 diabetic patients, and normal fibroblasts exposed chronically to high glucose, were defective in their capacity to up-regulate VEGF in response to hypoxia. In vivo, diabetic animals demonstrated an impaired ability to increase VEGF production in response to soft tissue ischemia. This resulted from a high glucose-induced decrease in transactivation by the transcription factor hypoxia-inducible factor-1alpha (HIF-1alpha), which mediates hypoxia-stimulated VEGF expression. Decreased HIF-1alpha functional activity was specifically caused by impaired HIF-1alpha binding to the coactivator p300. We identify covalent modification of p300 by the dicarbonyl metabolite methylglyoxal as being responsible for this decreased association. Administration of deferoxamine abrogated methylglyoxal conjugation, normalizing both HIF-1alpha/p300 interaction and transactivation by HIF-1alpha. In diabetic mice, deferoxamine promoted neovascularization and enhanced wound healing. These findings define molecular defects that underlie impaired VEGF production in diabetic tissues and offer a promising direction for therapeutic intervention.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/patología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Hipoxia/complicaciones , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Deferoxamina/farmacología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Glucosa/farmacología , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Neovascularización Patológica/complicaciones , Neovascularización Patológica/patología , Unión Proteica/efectos de los fármacos , Piruvaldehído/farmacología , Especies Reactivas de Oxígeno/metabolismo , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Factores de Transcripción p300-CBP/metabolismo
2.
Ann Surg ; 254(2): 217-25, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21606834

RESUMEN

OBJECTIVE: To test the hypothesis that the mechanical environment of cutaneous wounds can control scar formation. BACKGROUND: Mechanical forces have been recognized to modulate myriad biologic processes, but the role of physical force in scar formation remains unclear. Furthermore, the therapeutic benefits of offloading cutaneous wounds with a device have not been rigorously tested. METHODS: A mechanomodulating polymer device was utilized to manipulate the mechanical environment of closed cutaneous wounds in red Duroc swine. After 8 weeks, wounds subjected to different mechanical stress states underwent immunohistochemical analysis for fibrotic markers. In a phase I clinical study, 9 human patients undergoing elective abdominal surgery were treated postoperatively with a stress-shielding polymer on one side whereas the other side was treated as standard of care. Professional photographs were taken between 8 and 12 months postsurgery and evaluated using a visual analog scale by lay and professional panels. This study is registered with ClinicalTrials.gov, number NCT00766727. RESULTS: Stress shielding of swine incisions reduced histologic scar area by 6- and 9-fold compared to control and elevated stress states, respectively (P < 0.01 for both) and dramatically decreased the histologic expression of profibrotic markers. Closure of high-tension wounds induced human-like scar formation in the red Duroc, a phenotype effectively mitigated with stress shielding of wounds. In the study on humans, stress shielding of abdominal incisions significantly improved scar appearance (P = 0.004) compared with within-patient controls. CONCLUSIONS: These results indicate that mechanical manipulation of the wound environment with a dynamic stress-shielding polymer device can significantly reduce scar formation.


Asunto(s)
Cicatriz/fisiopatología , Modelos Animales de Enfermedad , Cicatrización de Heridas/fisiología , Pared Abdominal/patología , Pared Abdominal/fisiopatología , Pared Abdominal/cirugía , Adulto , Animales , Vendajes , Fenómenos Biomecánicos/fisiología , Cicatriz/patología , Cicatriz Hipertrófica/patología , Cicatriz Hipertrófica/fisiopatología , Cicatriz Hipertrófica/prevención & control , Fuerza Compresiva/fisiología , Femenino , Fibrosis , Humanos , Persona de Mediana Edad , Procedimientos de Cirugía Plástica , Factores de Riesgo , Elastómeros de Silicona , Piel/patología , Piel/fisiopatología , Dehiscencia de la Herida Operatoria/patología , Dehiscencia de la Herida Operatoria/fisiopatología , Porcinos , Adulto Joven
3.
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
4.
Stem Cells ; 27(1): 266-74, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18974212

RESUMEN

Evolving evidence suggests a possible role for adipose stromal cells (ASCs) in adult neovascularization, although the specific cues that stimulate their angiogenic behavior are poorly understood. We evaluated the effect of hypoxia, a central mediator of new blood vessel development within ischemic tissue, on proneovascular ASC functions. Murine ASCs were exposed to normoxia (21% oxygen) or hypoxia (5%, 1% oxygen) for varying lengths of time. Vascular endothelial growth factor (VEGF) secretion by ASCs increased as an inverse function of oxygen tension, with progressively higher VEGF expression at 21%, 5%, and 1% oxygen, respectively. Greater VEGF levels were also associated with longer periods in culture. ASCs were able to migrate towards stromal cell-derived factor (SDF)-1, a chemokine expressed by ischemic tissue, with hypoxia augmenting ASC expression of the SDF-1 receptor (CXCR4) and potentiating ASC migration. In vivo, ASCs demonstrated the capacity to proliferate in response to a hypoxic insult remote from their resident niche, and this was supported by in vitro studies showing increasing ASC proliferation with greater degrees of hypoxia. Hypoxia did not significantly alter the expression of endothelial surface markers by ASCs. However, these cells did assume an endothelial phenotype as evidenced by their ability to tubularize when seeded with differentiated endothelial cells on Matrigel. Taken together, these data suggest that ASCs upregulate their proneovascular activity in response to hypoxia, and may harbor the capacity to home to ischemic tissue and function cooperatively with existing vasculature to promote angiogenesis.


Asunto(s)
Tejido Adiposo/citología , Neovascularización Fisiológica , Células del Estroma/metabolismo , Animales , Hipoxia de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Citometría de Flujo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Fenotipo , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
FASEB J ; 23(3): 906-15, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19001054

RESUMEN

Classic tissue engineering paradigms are limited by the incorporation of a functional vasculature and a reliable means for reimplantation into the host circulation. We have developed a novel approach to overcome these obstacles using autologous explanted microcirculatory beds (EMBs) as bioscaffolds for engineering complex three-dimensional constructs. In this study, EMBs consisting of an afferent artery, capillary beds, efferent vein, and surrounding parenchymal tissue are explanted and maintained for 24 h ex vivo in a bioreactor that preserves EMB viability and function. Given the rapidly advancing field of stem cell biology, EMBs were subsequently seeded with three distinct stem cell populations, multipotent adult progenitor cells (MAPCs), and bone marrow and adipose tissue-derived mesenchymal stem cells (MSCs). We demonstrate MAPCs, as well as MSCs, are able to egress from the microcirculation into the parenchymal space, forming proliferative clusters. Likewise, human adipose tissue-derived MSCs were also found to egress from the vasculature and seed into the EMBs, suggesting feasibility of this technology for clinical applications. We further demonstrate that MSCs can be transfected to express a luciferase protein and continue to remain viable and maintain luciferase expression in vivo. By using the vascular network of EMBs, EMBs can be perfused ex vivo and seeded with stem cells, which can potentially be directed to differentiate into neo-organs or transfected to replace failing organs and deficient proteins.


Asunto(s)
Microcirculación/fisiología , Técnicas de Cultivo de Tejidos/métodos , Ingeniería de Tejidos/métodos , Animales , Reactores Biológicos , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Células Madre Mesenquimatosas/fisiología , Ratas , Ratas Desnudas , Piel/irrigación sanguínea , Transfección
7.
Organogenesis ; 10(2): 186-93, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24739276

RESUMEN

Cutaneous scarring is a major source of morbidity and current therapies to mitigate scar formation remain ineffective. Although wound fibrosis and inflammation are highly linked, only recently have mechanical forces been implicated in these pathways. Our group has developed a topical polymer device that significantly reduces post-injury scar formation via the manipulation of mechanical forces. Here we extend these studies to examine the genomewide transcriptional effects of mechanomodulation during scar formation using a validated large animal model, the red Duroc pig. We demonstrate that mechanical loading of incisional wounds upregulates expression of genes associated with inflammatory and fibrotic pathways, and that device-mediated offloading of these wounds reverses these effects. Validation studies are needed to clarify the clinical significance of these findings.


Asunto(s)
Regulación hacia Abajo/genética , Inflamación/genética , Transducción de Señal/genética , Transcripción Genética , Cicatrización de Heridas/genética , Heridas y Lesiones/genética , Animales , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Inflamación/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Sus scrofa , Heridas y Lesiones/patología
8.
Diabetes ; 63(9): 3047-56, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24740572

RESUMEN

Diabetic vascular pathology is largely attributable to impairments in tissue recovery from hypoxia. Circulating progenitor cells have been postulated to play a role in ischemic recovery, and deficiencies in these cells have been well described in diabetic patients. Here, we examine bone marrow-derived mesenchymal progenitor cells (BM-MPCs) that have previously been shown to be important for new blood vessel formation and demonstrate significant deficits in the context of diabetes. Further, we determine that this dysfunction is attributable to intrinsic defects in diabetic BM-MPCs that are not correctable by restoring glucose homeostasis. We identify two transcriptionally distinct subpopulations that are selectively depleted by both type 1 and type 2 diabetes, and these subpopulations have provasculogenic expression profiles, suggesting that they are vascular progenitor cells. These results suggest that the clinically observed deficits in progenitor cells may be attributable to selective and irreversible depletion of progenitor cell subsets in patients with diabetes.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Células Madre Mesenquimatosas , Células Madre/fisiología , Animales , Glucemia/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células Madre Mesenquimatosas/fisiología , Ratones , Parabiosis , Colgajos Quirúrgicos/irrigación sanguínea
9.
PLoS One ; 6(6): e21211, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731674

RESUMEN

An incomplete understanding of the nature of heterogeneity within stem cell populations remains a major impediment to the development of clinically effective cell-based therapies. Transcriptional events within a single cell are inherently stochastic and can produce tremendous variability, even among genetically identical cells. It remains unclear how mammalian cellular systems overcome this intrinsic noisiness of gene expression to produce consequential variations in function, and what impact this has on the biologic and clinical relevance of highly 'purified' cell subgroups. To address these questions, we have developed a novel method combining microfluidic-based single cell analysis and information theory to characterize and predict transcriptional programs across hundreds of individual cells. Using this technique, we demonstrate that multiple subpopulations exist within a well-studied and putatively homogeneous stem cell population, murine long-term hematopoietic stem cells (LT-HSCs). These subgroups are defined by nonrandom patterns that are distinguishable from noise and are consistent with known functional properties of these cells. We anticipate that this analytic framework can also be applied to other cell types to elucidate the relationship between transcriptional and phenotypic variation.


Asunto(s)
Células Madre Hematopoyéticas/citología , Teoría de la Información , Microfluídica/métodos , Análisis de la Célula Individual/métodos , Algoritmos , Animales , Análisis por Conglomerados , Citometría de Flujo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Análisis Multivariante , Factores de Tiempo , Transcripción Genética
10.
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
11.
Cell Cycle ; 9(1): 75-9, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20016290

RESUMEN

Diabetic wounds are a significant public health burden, with slow or nonhealing diabetic foot ulcers representing the leading cause of non-traumatic lower limb amputation in developed countries. These wounds heal poorly as a result of compromised blood vessel formation in response to ischemia. We have recently shown that this impairment in neovascularization results from a high glucose-induced defect in transactivation of hypoxia-inducible factor-1alpha (HIF-1alpha), the transcription factor regulating vascular endothelial growth factor (VEGF) expression. HIF-1 dysfunction is the end result of reactive oxygen species-induced modification of its coactivator p300 by the glycolytic metabolite methylglyoxal. Use of the iron chelator-antioxidant deferoxamine (DFO) reversed these effects and normalized healing of humanized diabetic wounds in mice. Here, we present additional data demonstrating that HIF-1alpha activity, not stability, is impaired in the high glucose environment. We demonstrate that high glucose-induced impairments in HIF-1alpha transactivation persist even in the setting of constitutive HIF-1alpha protein overexpression. Further, we show that high glucose-induced hydroxylation of the C-terminal transactivation domain of HIF-1alpha (the primary pathway regulating HIF-1alpha/p300 binding) does not alter HIF-1alpha activity. We extend our study of DFO's therapeutic efficacy in the treatment of impaired wound healing by demonstrating improvements in tissue viability in diabetic mice with DFO-induced increases in VEGF expression and vascular proliferation. Since DFO has been in clinical use for decades, the potential of this drug to treat a variety of ischemic conditions in humans can be evaluated relatively quickly.


Asunto(s)
Diabetes Mellitus/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Deferoxamina/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Humanos , Hipoxia/fisiopatología , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Sideróforos/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Plast Reconstr Surg ; 123(2 Suppl): 45S-55S, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19182663

RESUMEN

BACKGROUND: Cells from the bone marrow contribute to ischemic neovascularization, but the identity of these cells remains unclear. The authors identify mesenchymal stem cells as a bone marrow-derived progenitor population that is able to engraft into peripheral tissue in response to ischemia. METHODS: A murine model of skin ischemia was used. Bone marrow, blood, and skin were harvested at different time points and subjected to flow cytometric analysis for mesenchymal and hematopoietic markers (n = 3 to 7 per time point). Using a parabiotic model pairing donor green fluorescent protein (GFP)-positive with recipient wild-type mice, progenitor cell engraftment was examined in ischemic tissue by fluorescence microscopy, and engrafted cells were analyzed by flow cytometry for endothelial and mesenchymal markers. In vitro, the ability of both bone marrow- and adipose-derived mesenchymal stem cells to adopt endothelial characteristics was examined by analyzing (1) the ability of mesenchymal stem cells to take up DiI-acetylated low-density lipoprotein and Alexa Fluor lectin, and (2) phenotypic changes of mesenchymal stem cells co-cultured with GFP-labeled endothelial cells or under hypoxic/vascular endothelial growth factor stimulation. RESULTS: In vivo, the bone marrow mesenchymal stem cell population decreased significantly immediately after surgery, with subsequent engraftment of these cells in ischemic tissue. Engrafted cells lacked the panhematopoietic antigen CD45, consistent with a mesenchymal origin. In vitro, bone marrow- and adipose-derived mesenchymal stem cells took up DiI-acetylated low-density lipoprotein and Alexa Fluor lectin, and expressed endothelial markers under hypoxic conditions. CONCLUSIONS: The authors' data suggest that mesenchymal precursor cells can give rise to endothelial progenitors. Consequently, cell-based therapies augmenting the mesenchymal stem cell population could represent powerful alternatives to current therapies for ischemic vascular disease.


Asunto(s)
Isquemia/terapia , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica/fisiología , Piel/irrigación sanguínea , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular , Células Endoteliales/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Animales
14.
Plast Reconstr Surg ; 123(2 Suppl): 65S-75S, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19182665

RESUMEN

BACKGROUND: Age-related impairments in wound healing are associated with decreased neovascularization, a process that is regulated by hypoxia-responsive cytokines, including stromal cell-derived factor (SDF)-1 alpha. Interleukin-1 beta is an important inflammatory cytokine involved in wound healing and is believed to regulate SDF-1 alpha expression independent of hypoxia signaling. Thus, the authors examined the relative importance of interleukin (IL)-1 beta and hypoxia-inducible factor (HIF)-1 alpha on SDF-1 alpha expression in aged wound healing. METHODS: Young and aged mice (n = 4 per group) were examined for wound healing using a murine excisional wound model. Wounds were harvested at days 0, 1, 3, 5, and 7 for histologic analysis, immunohistochemistry, enzyme-linked immunosorbent assay, and Western blot. An engineered wild-type and mutated SDF luciferase reporter construct were used to determine HIF transactivation. RESULTS: Aged mice demonstrated significantly impaired wound healing, reduced granulation tissue, and increased epithelial gap compared with young controls. Real-time polymerase chain reaction demonstrated reduced SDF-1 alpha levels in aged wounds that correlated with reduced CD31+ neovessels. Western blots revealed decreased HIF-1 alpha protein in aged wounds. However, both IL-1 beta and macrophage infiltrate were unchanged between young and aged animals. Using the wild-type and mutated SDF luciferase reporter construct in which the hypoxia response element was deleted, only young fibroblasts were able to respond to IL-1 beta stimulation, and this response was abrogated by mutating the HIF-binding sites. This suggests that HIF binding is essential for SDF-1 transactivation in response to both inflammatory and hypoxic stimuli. CONCLUSIONS: SDF-1 alpha deficiency observed during aged wound healing is attributable predominantly to decreased HIF-1 alpha levels rather than impaired IL-1 beta expression.


Asunto(s)
Envejecimiento/fisiología , Quimiocina CXCL12/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Cicatrización de Heridas/fisiología , Animales , Quimiocina CXCL12/deficiencia , Modelos Animales de Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA