Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(34): 13785-90, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23918395

RESUMO

Endothelial cells play significant roles in conditioning tissues after injury by the production and secretion of angiocrine factors. At least two distinct subsets of monocytes, CD45(+)CD11b(+)Gr1(+)Ly6C(+) inflammatory and CD45(+)CD11b(+)Gr1(-)Ly6C(-) anti-inflammatory monocytes, respond differentially to these angiocrine factors and promote pathogen/debris clearance and arteriogenesis/tissue regeneration, respectively. We demonstrate here that local sphingosine 1-phosphate receptor 3 (S1P3) agonism recruits anti-inflammatory monocytes to remodeling vessels. Poly(lactic-co-glycolic acid) thin films were used to deliver FTY720, an S1P1/3 agonist, to inflamed and ischemic tissues, which resulted in a reduction in proinflammatory cytokine secretion and an increase in regenerative cytokine secretion. The altered balance of cytokine secretion results in preferential recruitment of anti-inflammatory monocytes from circulation. The chemotaxis of these cells, which express more S1P3 than inflammatory monocytes, toward SDF-1α was also enhanced with FTY720 treatment, but not in S1P3 knockout cells. FTY720 delivery enhanced arteriolar diameter expansion and increased length density of the local vasculature. This work establishes a role for S1P receptor signaling in the local conditioning of tissues by angiocrine factors that preferentially recruit regenerative monocytes that can enhance healing outcomes, tissue regeneration, and biomaterial implant functionality.


Assuntos
Monócitos/fisiologia , Neovascularização Fisiológica/fisiologia , Propilenoglicóis/farmacologia , Próteses e Implantes/efeitos adversos , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Engenharia Tecidual/métodos , Lesões do Sistema Vascular/tratamento farmacológico , Análise de Variância , Animais , Western Blotting , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Citocinas/metabolismo , Primers do DNA/genética , Portadores de Fármacos , Cloridrato de Fingolimode , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Ácido Láctico , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microvasos/citologia , Monócitos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Propilenoglicóis/administração & dosagem , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Lisoesfingolipídeo/agonistas , Esfingosina/administração & dosagem , Esfingosina/farmacologia , Lesões do Sistema Vascular/etiologia
2.
PLoS One ; 14(7): e0219254, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31276542

RESUMO

Thermoresponsive polymers, such as poly(N-isopropyl acrylamide) (PNIPAM), have been identified and used as cell culture substrates, taking advantage of the polymer's lower critical solution temperature (LCST) to mechanically harvest cells. This technology bypasses the use of biochemical enzymes that cleave important cell-cell and cell-matrix interactions. In this study, the process of electrospinning is used to fabricate and characterize aligned PNIPAM nanofiber scaffolds that are biocompatible and thermoresponsive. Nanofiber scaffolds produced by electrospinning possess a 3D architecture that mimics native extracellular matrix, providing physical and chemical cues to drive cell function and phenotype. We present a factorial design of experiments (DOE) approach to systematically determine the effects of different electrospinning process parameters on PNIPAM nanofiber diameter and alignment. Results show that high molecular weight PNIPAM can be successfully electrospun into both random and uniaxially aligned nanofiber mats with similar fiber diameters by simply altering the speed of the rotating mandrel collector from 10,000 to 33,000 RPM. PNIPAM nanofibers were crosslinked with OpePOSS, which was verified using FTIR. The mechanical properties of the scaffolds were characterized using dynamic mechanical analysis, revealing an order of magnitude difference in storage modulus (MPa) between cured and uncured samples. In summary, cross-linked PNIPAM nanofiber scaffolds were determined to be stable in aqueous culture, biocompatible, and thermoresponsive, enabling their use in diverse cell culture applications.


Assuntos
Resinas Acrílicas/química , Nanofibras/química , Engenharia Tecidual/métodos , Acrilamidas/química , Materiais Biocompatíveis/química , Matriz Extracelular , Polímeros/química , Alicerces Teciduais/química
3.
Biomaterials ; 29(19): 2869-77, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18405965

RESUMO

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.


Assuntos
Artérias/crescimento & desenvolvimento , Desenvolvimento Ósseo , Lisofosfolipídeos/administração & dosagem , Esfingosina/análogos & derivados , Engenharia Tecidual , Animais , Bromodesoxiuridina , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Esfingosina/administração & dosagem
4.
J Biomed Mater Res A ; 105(9): 2416-2428, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28467030

RESUMO

Thermoresponsive polymer (TRP) cell culture substrates are widely utilized for nonenzymatic, temperature-triggered release of adherent cells. Increasingly, multicomponent TRPs are being developed to facilitate refined control of cell adhesion and detachment, which requires an understanding of the relationships between composition-dependent substrate physicochemical properties and cellular responses. Here, we utilize a homologous series of poly(MEO2 MAx -co-OEGMAy ) brushes with variable copolymer ratio (x/y) to explore the effects of substrate hydrophobicity on L-929 fibroblast adhesion, morphology, and temperature-triggered cell detachment. Substrate hydrophobicity is reported in terms of the equilibrium spreading coefficient (S), and variations in copolymer ratio reveal differential hydrophobicity that is correlated to serum protein adsorption and initial cell attachment at 37°C. Furthermore, quantitative metrics of cell morphology show that cell spreading is enhanced on more hydrophobic surfaces with increased (x/y) ratio, which is further supported by gene expression analysis of biomarkers of cell spreading (e.g., RhoA, Dusp2). Temperature-dependent cell detachment is limited for pure poly(MEO2 MA); however, rapid cell rounding and detachment (<20 min) are evident for all poly(MEO2 MAx -co-OEGMAy ) substrates. These results suggest that increased MEO2 MA content in poly(MEO2 MAx -co-OEGMAy ) substrates elicits enhanced protein adsorption, cell adhesion, and cell spreading; however, integration of small amounts of the more hydrophilic OEGMA unit facilitates both cell attachment/spreading and detachment. This study demonstrates an important role for the composition-dependent control of surface hydrophobicity in the design of multicomponent TRPs for desired biological outcomes. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2416-2428, 2017.


Assuntos
Interações Hidrofóbicas e Hidrofílicas , Metacrilatos/farmacologia , Polímeros/farmacologia , Temperatura , Adsorção , Animais , Proteínas Sanguíneas/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Forma Celular/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Cinética , Camundongos
5.
ACS Appl Mater Interfaces ; 7(4): 2518-28, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25569588

RESUMO

Adhesion control in liquid-liquid-solid systems represents a challenge for applications ranging from self-cleaning to biocompatibility of engineered materials. By using responsive polymer chemistry and molecular self-assembly, adhesion at solid/liquid interfaces can be achieved and modulated by external stimuli. Here, we utilize thermosensitive polymeric materials based on random copolymers of di(ethylene glycol) methyl ether methacrylate (x = MEO2MA) and oligo(ethylene glycol) methyl ether methacrylate (y = OEGMA), that is, P(MEO2MAx-co-OEGMAy), to investigate the role of hydrophobicity on the phenomenon of adhesion. The copolymer ratio (x/y) dictates macromolecular changes enabling control of the hydrophilic-to-lipophilic balance (HBL) of the polymer brushes through external triggers such as ionic strength and temperature. We discuss the HBL of the thermobrushes in terms of the surface energy of the substrate by measuring the contact angle at water-decane-P(MEO2MAx-co-OEGMAy) brush contact line as a function of polymer composition and temperature. Solid supported polyelectrolyte layers grafted with P(MEO2MAx-co-OEGMAy) display a transition in the wettability that is related to the lower critical solution temperature of the polymer brushes. Using experimental observation of the hydrophilic to hydrophobic transition by the contact angle, we extract the underlying energetics associated with liquid-liquid-solid adhesion as a function of the copolymer ratio. The change in cellular attachment on P(MEO2MAx-co-OEGMAy) substrates of variable (x/y) composition demonstrates the subtle role of compositional tuning on the ability to control liquid-liquid-solid adhesion in biological applications.


Assuntos
Técnicas de Cultura de Células/instrumentação , Polímeros/química , Animais , Adesão Celular , Fibroblastos/fisiologia , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Temperatura , Molhabilidade
6.
Acta Biomater ; 10(11): 4704-4714, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25128750

RESUMO

Biomaterial-mediated controlled release of soluble signaling molecules is a tissue engineering approach to spatially control processes of inflammation, microvascular remodeling and host cell recruitment, and to generate biochemical gradients in vivo. Lipid mediators, such as sphingosine 1-phosphate (S1P), are recognized for their essential roles in spatial guidance, signaling and highly regulated endogenous gradients. S1P and pharmacological analogs such as FTY720 are therapeutically attractive targets for their critical roles in the trafficking of cells between blood and tissue spaces, both physiologically and pathophysiologically. However, the interaction of locally delivered sphingolipids with the complex metabolic networks controlling the flux of lipid species in inflamed tissue has yet to be elucidated. In this study, complementary in vitro and in vivo approaches are investigated to identify relationships between polymer composition, drug release kinetics, S1P metabolic activity, signaling gradients and spatial positioning of circulating cells around poly(lactic-co-glycolic acid) biomaterials. Results demonstrate that biomaterial-based gradients of S1P are short-lived in the tissue due to degradation by S1P lyase, an enzyme that irreversibly degrades intracellular S1P. On the other hand, in vivo gradients of the more stable compound, FTY720, enhance microvascular remodeling by selectively recruiting an anti-inflammatory subset of monocytes (S1P3(high)) to the biomaterial. Results highlight the need to better understand the endogenous balance of lipid import/export machinery and lipid kinase/phosphatase activity in order to design biomaterial products that spatially control the innate immune environment to maximize regenerative potential.


Assuntos
Inflamação/patologia , Microvasos/patologia , Microvasos/fisiopatologia , Receptores de Lisoesfingolipídeo/metabolismo , Engenharia Tecidual/métodos , Remodelação Vascular , Animais , Cloridrato de Fingolimode , Cinética , Ácido Láctico/química , Ligantes , Linfócitos/efeitos dos fármacos , Lisofosfolipídeos , Masculino , Camundongos Endogâmicos C57BL , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Propilenoglicóis , Próteses e Implantes , Receptores de Lisoesfingolipídeo/agonistas , Esfingolipídeos/metabolismo , Esfingosina/análogos & derivados
7.
Tissue Eng Part A ; 17(5-6): 617-29, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20874260

RESUMO

Proper spatial and temporal regulation of microvascular remodeling is critical to the formation of functional vascular networks, spanning the various arterial, venous, capillary, and collateral vessel systems. Recently, our group has demonstrated that sustained release of sphingosine 1-phosphate (S1P) from biodegradable polymers promotes microvascular network growth and arteriolar expansion. In this study, we employed S1P receptor-specific compounds to activate and antagonize different combinations of S1P receptors to elucidate those receptors most critical for promotion of pharmacologically induced microvascular network growth. We show that S1P(1) and S1P(3) receptors act synergistically to enhance functional network formation via increased functional length density, arteriolar diameter expansion, and increased vascular branching in the dorsal skinfold window chamber model. FTY720, a potent activator of S1P(1) and S1P(3), promoted a 107% and 153% increase in length density 3 and 7 days after implantation, respectively. It also increased arteriolar diameters by 60% and 85% 3 and 7 days after implantation. FTY720-stimulated branching in venules significantly more than unloaded poly(D, L-lactic-co-glycolic acid). When implanted on the mouse spinotrapezius muscle, FTY720 stimulated an arteriogenic response characterized by increased tortuosity and collateralization of branching microvascular networks. Our results demonstrate the effectiveness of S1P(1) and S1P(3) receptor-selective agonists (such as FTY720) in promoting microvascular growth for tissue engineering applications.


Assuntos
Microvasos/crescimento & desenvolvimento , Microvasos/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Actinas/metabolismo , Animais , Preparações de Ação Retardada , Cloridrato de Fingolimode , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/efeitos dos fármacos , Modelos Animais , Músculos/efeitos dos fármacos , Músculos/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/química , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia
8.
Tissue Eng Part B Rev ; 16(3): 361-70, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20121415

RESUMO

Microvascular remodeling is a complex process that includes many cell types and molecular signals. Despite a continued growth in the understanding of signaling pathways involved in the formation and maturation of new blood vessels, approximately half of all compounds entering clinical trials will fail, resulting in the loss of much time, money, and resources. Most pro-angiogenic clinical trials to date have focused on increasing neovascularization via the delivery of a single growth factor or gene. Alternatively, a focus on the concerted regulation of whole networks of genes may lead to greater insight into the underlying physiology since the coordinated response is greater than the sum of its parts. Systems biology offers a comprehensive network view of the processes of angiogenesis and arteriogenesis that might enable the prediction of drug targets and whether or not activation of the targets elicits the desired outcome. Systems biology integrates complex biological data from a variety of experimental sources (-omics) and analyzes how the interactions of the system components can give rise to the function and behavior of that system. This review focuses on how systems biology approaches have been applied to microvascular growth and remodeling, and how network analysis tools can be utilized to aid novel pro-angiogenic drug discovery.


Assuntos
Microvasos/fisiologia , Biologia de Sistemas/métodos , Engenharia Tecidual/métodos , Prótese Vascular , Regeneração Tecidual Guiada/métodos , Humanos , Modelos Biológicos
9.
Biomaterials ; 31(25): 6417-24, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20621764

RESUMO

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.


Assuntos
Transplante Ósseo/fisiologia , Imunossupressores/administração & dosagem , Propilenoglicóis/administração & dosagem , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Remodelação Óssea , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/ultraestrutura , Materiais Revestidos Biocompatíveis/química , Força Compressiva , Módulo de Elasticidade , Cloridrato de Fingolimode , Imunossupressores/uso terapêutico , Masculino , Neovascularização Fisiológica , Osseointegração , Propilenoglicóis/uso terapêutico , Ratos , Ratos Sprague-Dawley , Esfingosina/administração & dosagem , Esfingosina/uso terapêutico , Engenharia Tecidual , Transplante Homólogo
10.
Tissue Eng Part A ; 16(6): 1801-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20038198

RESUMO

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.


Assuntos
Doenças Ósseas/cirurgia , Regeneração Óssea/efeitos dos fármacos , Imunossupressores/farmacologia , Microvasos/efeitos dos fármacos , Propilenoglicóis/farmacologia , Crânio/cirurgia , Esfingosina/análogos & derivados , Alicerces Teciduais/química , Animais , Doenças Ósseas/diagnóstico por imagem , Doenças Ósseas/tratamento farmacológico , Cloridrato de Fingolimode , Imunossupressores/química , Imunossupressores/uso terapêutico , Masculino , Microvasos/diagnóstico por imagem , Propilenoglicóis/química , Propilenoglicóis/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/agonistas , Crânio/diagnóstico por imagem , Crânio/efeitos dos fármacos , Crânio/patologia , Esfingosina/química , Esfingosina/farmacologia , Esfingosina/uso terapêutico , Microtomografia por Raio-X
11.
Tissue Eng Part C Methods ; 15(1): 11-21, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18844601

RESUMO

The basement membrane protein, laminin I, has been used broadly as a planar two-dimensional film or in a three-dimensional form as a reconstituted basement membrane gel such as Matrigel to support cellular attachment, growth, and differentiation in vitro. In basement membranes in vivo, laminin exhibits a fibrillar morphology, highlighting the electrospinning process as an ideal method to recreate such fibrous substrates in vitro. Electrospinning was employed to fabricate meshes of murine laminin I nanofibers (LNFs) with fiber size, geometry, and porosity of authentic basement membranes. Purified laminin I was solubilized and electrospun in parametric studies of fiber diameters as a function of polymer solution concentration, collecting distance, and flow rate. Resulting fiber diameters ranged from 90 to 300 nm with mesh morphologies containing beads. Unlike previously described nanofibers (NFs) synthesized from proteins such as collagen, meshes of LNFs retain their structural features when wetted and do not require fixation by chemical crosslinking, which often destroys cell attachment and other biological activity. The LNF meshes maintained their geometry for at least 2 days in culture without chemical crosslinking. PC12 cells extended neurites without nerve growth factor stimulation on LNF substrates. Additionally, LNFs significantly enhance both the rate and quantity of attachment of human adipose stem cells (ASCs) compared to laminin films. ASCs were viable and maintained attachment to LNF meshes in serum-free media for at least 3 days in culture and extended neurite-like processes after 24 h in serum-free media conditions without media additives to induce differentiation. LNF meshes are a novel substrate for cell studies in vitro, whose properties may be an excellent scaffold material for delivering cells in tissue engineering applications in vivo.


Assuntos
Membrana Basal/metabolismo , Laminina/metabolismo , Nanoestruturas/química , Tecido Adiposo/citologia , Animais , Membrana Basal/efeitos dos fármacos , Meios de Cultura , Humanos , Laminina/ultraestrutura , Nanoestruturas/ultraestrutura , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Células PC12 , Ratos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/ultraestrutura
12.
Organogenesis ; 4(4): 215-27, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19337401

RESUMO

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.

13.
J Tissue Eng Regen Med ; 2(4): 210-20, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18493910

RESUMO

Electrospinning has recently gained widespread attention as a process capable of producing nanoscale fibres that mimic native extracellular matrix. In this study, we compared the osteogenic differentiation behaviour of human adipose stem cells (ASCs) on a 3D nanofibre matrix of type I rat tail collagen (RTC) and a 2D RTC collagen-coated substrate, using a novel serum-free osteogenic medium. The serum-free medium significantly enhanced the numbers of proliferating cells in culture, compared to ASCs in traditional basal medium containing 10% animal serum, highlighting a potential clinical role for in vitro stem cell expansion. Osteogenic differentiation behaviour was assessed at days 7, 14 and 21 using quantitative real-time RT-PCR analysis of the osteogenic genes collagen I (Coll I), alkaline phosphatase (ALP), osteopontin (OP), osteonectin (ON), osteocalcin (OC) and core-binding factor-alpha (cbfa1). All genes were upregulated (>one-fold) in ASCs cultured on nanofibre scaffolds over 2D collagen coatings by day 21. Synthesis of mineralized extracellular matrix on the scaffolds was assessed on day 21 with Alizarin red staining. These studies demonstrate that 3D nanoscale morphology plays a critical role in regulating cell fate processes and in vitro osteogenic differentiation of ASCs under serum-free conditions.


Assuntos
Adipócitos/citologia , Materiais Biomiméticos/metabolismo , Diferenciação Celular , Colágeno/metabolismo , Nanoestruturas , Osteogênese , Células-Tronco/citologia , Adulto , Animais , Antraquinonas , Ciclo Celular , Forma Celular , Sobrevivência Celular , Células Cultivadas , Colágeno/ultraestrutura , Meios de Cultura Livres de Soro , Feminino , Regulação da Expressão Gênica , Humanos , Nanoestruturas/ultraestrutura , Osteogênese/genética , Pseudópodes , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Biomaterials ; 29(35): 4698-708, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18804278

RESUMO

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.


Assuntos
Indóis/farmacologia , Microvasos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Receptores CCR2/deficiência , Receptores CCR2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA