Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Immunity ; 47(5): 862-874.e3, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166587

RESUMEN

Chemoattractant-mediated recruitment of hematopoietic cells to sites of pathogen growth or tissue damage is critical to host defense and organ homeostasis. Chemotaxis is typically considered to rely on spatial sensing, with cells following concentration gradients as long as these are present. Utilizing a microfluidic approach, we found that stable gradients of intermediate chemokines (CCL19 and CXCL12) failed to promote persistent directional migration of dendritic cells or neutrophils. Instead, rising chemokine concentrations were needed, implying that temporal sensing mechanisms controlled prolonged responses to these ligands. This behavior was found to depend on G-coupled receptor kinase-mediated negative regulation of receptor signaling and contrasted with responses to an end agonist chemoattractant (C5a), for which a stable gradient led to persistent migration. These findings identify temporal sensing as a key requirement for long-range myeloid cell migration to intermediate chemokines and provide insights into the mechanisms controlling immune cell motility in complex tissue environments.


Asunto(s)
Movimiento Celular , Factores Quimiotácticos/fisiología , Células Mieloides/fisiología , Animales , Quimiocina CCL19/fisiología , Quimiocina CXCL12/fisiología , Células Dendríticas/fisiología , Quinasa 3 del Receptor Acoplado a Proteína-G/fisiología , Quinasas de Receptores Acoplados a Proteína-G/fisiología , Ratones , Ratones Endogámicos C57BL , Microfluídica
2.
Birth Defects Res C Embryo Today ; 90(1): 67-74, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20301222

RESUMEN

Adult mesenchymal stem cells (MSCs) include a select population of resident cells within adult tissues, which retain the ability to differentiate along several tissue-specific lineages under defined media conditions and have finite expansion potential in vitro. These adult progenitor populations have been identified in various tissues, but it remains unclear exactly what role both transplanted and native MSCs play in processes of disease and regeneration. Interestingly, increasing evidence reveals a unique antiinflammatory immunomodulatory phenotype shared among this population, lending support to the idea that MSCs play a central role in early tissue remodeling responses where a controlled inflammatory response is required. However, additional evidence suggests that MSCs may not retain infinite immune privilege and that the context with which these cells are introduced in vivo may influence their immune phenotype. Therefore, understanding this dynamic microenvironment in which MSCs participate in complex feedback loops acting upon and being influenced by a plethora of secreted cytokines, extracellular matrix molecules, and fragments will be critical to elucidating the role of MSCs in the intertwined processes of immunomodulation and tissue repair.


Asunto(s)
Células Madre Adultas/inmunología , Inmunomodulación/fisiología , Células Madre Mesenquimatosas/inmunología , Animales , Diferenciación Celular , Citocinas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Terapia Genética , Humanos , Inflamación/inmunología , Regeneración/fisiología , Trasplante de Células Madre
3.
Biomaterials ; 29(19): 2869-77, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18405965

RESUMEN

Sphingosine 1-phosphate (S1P) is a bioactive phospholipid that impacts migration, proliferation, and survival in diverse cell types, including endothelial cells, smooth muscle cells, and osteoblast-like cells. In this study, we investigated the effects of sustained release of S1P on microvascular remodeling and associated bone defect healing in vivo. The murine dorsal skinfold window chamber model was used to evaluate the structural remodeling response of the microvasculature. Our results demonstrated that 1:400 (w/w) loading and subsequent sustained release of S1P from poly(lactic-co-glycolic acid) (PLAGA) significantly enhanced lumenal diameter expansion of arterioles and venules after 3 and 7 days. Incorporation of 5-bromo-2-deoxyuridine (BrdU) at day 7 revealed significant increases in mural cell proliferation in response to S1P delivery. Additionally, three-dimensional (3D) scaffolds loaded with S1P (1:400) were implanted into critical-size rat calvarial defects, and healing of bony defects was assessed by radiograph X-ray, microcomputed tomography (muCT), and histology. Sustained release of S1P significantly increased the formation of new bone after 2 and 6 weeks of healing and histological results suggest increased numbers of blood vessels in the defect site. Taken together, these experiments support the use of S1P delivery for promoting microvessel diameter expansion and improving the healing outcomes of tissue-engineered therapies.


Asunto(s)
Arterias/crecimiento & desarrollo , Desarrollo Óseo , Lisofosfolípidos/administración & dosificación , Esfingosina/análogos & derivados , Ingeniería de Tejidos , Animales , Bromodesoxiuridina , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Esfingosina/administración & dosificación
4.
Laryngoscope ; 120(5): 895-901, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20422682

RESUMEN

OBJECTIVES/HYPOTHESIS: Advances in bone repair have focused on the minimally-invasive delivery of tissue-engineered bone (TEB). A promising injectable biopolymer of chitosan and inorganic phosphates was seeded with mesenchymal stem cells (MSCs) and a bone growth factor (BMP-2), and evaluated in a rat calvarial critical size defect (CSD). Green fluorescent protein (GFP)-labeled MSCs are used to evaluate patterns of cell viability and proliferation. STUDY DESIGN: Prospective, controlled trial in an animal model. METHODS: In 30 male rats, 8-mm calvarial CSDs were created, and divided into five groups of six animals each. In the experimental groups, the defects were injected with either chitosan gel, gel loaded with MSCs (0.3 x 10(6) cells/defect), gel loaded with BMP-2 (2 microg/defect), or gel loaded with both MSC and BMP-2. In the control group, the defect was left untreated. At 4 weeks, in vivo microcomputed tomography (micro-CT) analysis was performed. At 8 weeks, calvarial specimens were examined by micro-CT, histology, and immunohistochemistry. RESULTS: New areas of bone growth were seen in the defects of all treated animals. Micro-CT analysis revealed a significant (P < .001) time-dependent increase in the regeneration of bone volume and bone area in defects treated with gel/MSC/BMP-2 as compared to all other groups. Histological analysis confirmed this difference. GFP-labeled TEB was detected within the areas of new bone, indicating cell viability and contribution to new bone growth by the injected MSC. CONCLUSIONS: This study demonstrates that an injectable form of TEB using a chitosan gel, MSC, and BMP-2 can enhance bone formation in a rat calvarial CSD.


Asunto(s)
Proteína Morfogenética Ósea 2/administración & dosificación , Trasplante de Células Madre Mesenquimatosas/métodos , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Procedimientos de Cirugía Plástica/métodos , Cráneo/cirugía , Ingeniería de Tejidos/métodos , Animales , Regeneración Ósea/fisiología , Quitosano , Geles , Procesamiento de Imagen Asistido por Computador , Inyecciones , Masculino , Fosfatos , Ratas , Ratas Sprague-Dawley , Cráneo/patología , Microtomografía por Rayos X
5.
Tissue Eng Part A ; 16(6): 1801-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20038198

RESUMEN

The calvarial bone microenvironment contains a unique progenitor niche that should be considered for therapeutic manipulation when designing regeneration strategies. Recently, our group demonstrated that cells isolated from the dura are multipotent and exhibit expansion potential and robust mineralization on biodegradable constructs in vitro. In this study, we evaluate the effectiveness of healing critical-sized cranial bone defects by enhancing microvascular network growth and host dura progenitor trafficking to the defect space pharmacologically by delivering drugs targeted to sphingosine 1-phosphate (S1P) receptors. We demonstrate that delivery of pharmacological agonists to (S1P) receptors S1P(1) and S1P(3) significantly increase bone ingrowth, total microvessel density, and smooth muscle cell investment on nascent microvessels within the defect space. Further, in vitro proliferation and migration studies suggest that selective activation of S1P(3) promotes recruitment and growth of osteoblastic progenitors from the meningeal dura mater.


Asunto(s)
Enfermedades Óseas/cirugía , Regeneración Ósea/efectos de los fármacos , Inmunosupresores/farmacología , Microvasos/efectos de los fármacos , Glicoles de Propileno/farmacología , Cráneo/cirugía , Esfingosina/análogos & derivados , Andamios del Tejido/química , Animales , Enfermedades Óseas/diagnóstico por imagen , Enfermedades Óseas/tratamiento farmacológico , Clorhidrato de Fingolimod , Inmunosupresores/química , Inmunosupresores/uso terapéutico , Masculino , Microvasos/diagnóstico por imagen , Glicoles de Propileno/química , Glicoles de Propileno/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/agonistas , Cráneo/diagnóstico por imagen , Cráneo/efectos de los fármacos , Cráneo/patología , Esfingosina/química , Esfingosina/farmacología , Esfingosina/uso terapéutico , Microtomografía por Rayos X
6.
Biomaterials ; 31(25): 6417-24, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20621764

RESUMEN

Poor vascularization coupled with mechanical instability is the leading cause of post-operative complications and poor functional prognosis of massive bone allografts. To address this limitation, we designed a novel continuous polymer coating system to provide sustained localized delivery of pharmacological agent, FTY720, a selective agonist for sphingosine 1-phosphate receptors, within massive tibial defects. In vitro drug release studies validated 64% loading efficiency with complete release of compound following 14 days. Mechanical evaluation following six weeks of healing suggested significant enhancement of mechanical stability in FTY720 treatment groups compared with unloaded controls. Furthermore, superior osseous integration across the host-graft interface, significant enhancement in smooth muscle cell investment, and reduction in leukocyte recruitment was evident in FTY720 treated groups compared with untreated groups. Using this approach, we can capitalize on the existing mechanical and biomaterial properties of devitalized bone, add a controllable delivery system while maintaining overall porous structure, and deliver a small molecule compound to constitutively target vascular remodeling, osseous remodeling, and minimize fibrous encapsulation within the allograft-host bone interface. Such results support continued evaluation of drug-eluting allografts as a viable strategy to improve functional outcome and long-term success of massive cortical allograft implants.


Asunto(s)
Trasplante Óseo/fisiología , Inmunosupresores/administración & dosificación , Glicoles de Propileno/administración & dosificación , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Remodelación Ósea , Huesos/efectos de los fármacos , Huesos/ultraestructura , Materiales Biocompatibles Revestidos/química , Fuerza Compresiva , Módulo de Elasticidad , Clorhidrato de Fingolimod , Inmunosupresores/uso terapéutico , Masculino , Neovascularización Fisiológica , Oseointegración , Glicoles de Propileno/uso terapéutico , Ratas , Ratas Sprague-Dawley , Esfingosina/administración & dosificación , Esfingosina/uso terapéutico , Ingeniería de Tejidos , Trasplante Homólogo
7.
J Biomed Mater Res A ; 89(3): 632-41, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18442122

RESUMEN

Bony craniofacial deficits resulting from injury, disease, or birth defects remain a considerable clinical challenge. In this study, microsphere-based scaffold fabrication methods were use to study the respective effects of scaffold pore size, open pore volume, and total void volume fraction on osseous tissue infiltration and bone regeneration in a critical size rat cranial defect. To compare the healing effects of these parameters, three different scaffolds types were fabricated: solid 100 microm spheres, solid 500 microm spheres, and hollow 500 microm spheres. These constructs were implanted into surgically created rat calvarial defects. By 90-days post op, results of micro computed tomography (CT) analysis showed that all scaffolds generated similar amounts of new bone which was significantly greater than untreated controls. Interestingly, the spatial distribution of new bone within the defect area varied by scaffold group. MicroCT and histological analysis demonstrated healing restricted to the dural side in the hollow 500 microm group, whereas the solid 500 microm group demonstrated healing along the dural side and within the center of the defect. Solid 100 microm groups demonstrated healing along the dural layer, periosteal layer, and within the center of the defect. These results suggest that pore size and closed void volume may both play important roles in scaffold degradation patterns and associated bone healing.


Asunto(s)
Microesferas , Cráneo/patología , Andamios del Tejido , Cicatrización de Heridas , Animales , Porosidad , Ratas , Cráneo/diagnóstico por imagen , Microtomografía por Rayos X
8.
Organogenesis ; 4(4): 215-27, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19337401

RESUMEN

Vascular growth and remodeling are complex processes that depend on the proper spatial and temporal regulation of many different signaling molecules to form functional vascular networks. The ability to understand and regulate these signals is an important clinical need with the potential to treat a wide variety of disease pathologies. Current approaches have focused largely on the delivery of proteins to promote neovascularization of ischemic tissues, most notably VEGF and FGF. Although great progress has been made in this area, results from clinical trials are disappointing and safer and more effective approaches are required. To this end, biological agents used for therapeutic neovascularization must be explored beyond the current well-investigated classes. This review focuses on potential pathways for novel drug discovery, utilizing small molecule approaches to induce and enhance neovascularization. Specifically, four classes of new and existing molecules are discussed, including transcriptional activators, receptor selective agonists and antagonists, natural product-derived small molecules, and novel synthetic small molecules.

9.
Biomaterials ; 29(35): 4698-708, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18804278

RESUMEN

Phthalimide neovascular factor (PNF1, formerly SC-3-149) is a potent stimulator of proangiogenic signaling pathways in endothelial cells. In this study, we evaluated the in vivo effects of sustained PNF1 release to promote ingrowth and expansion of microvascular networks surrounding biomaterial implants. The dorsal skinfold window chamber was used to evaluate the structural remodeling response of the local microvasculature. PNF1 was released from poly(lactic-co-glycolic acid) (PLAGA) films, and a transport model was utilized to predict PNF1 penetration into the surrounding tissue. PNF1 significantly expanded microvascular networks within a 2mm radius from implants after 3 and 7 days by increasing microvessel length density and lumenal diameter of local arterioles and venules. Staining of histological sections with CD11b showed enhanced recruitment of circulating white blood cells, including monocytes, which are critical for the process of vessel enlargement through arteriogenesis. As PNF1 has been shown to modulate MT1-MMP, a facilitator of CCL2 dependent leukocyte transmigration, aspects of window chamber experiments were repeated in CCR2(-/-) (CCL2 receptor) mouse chimeras to more fully explore the critical nature of monocyte recruitment on the therapeutic benefits of PNF1 function in vivo.


Asunto(s)
Indoles/farmacología , Microvasos/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Receptores CCR2/deficiencia , Receptores CCR2/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda