Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biomed Mater Res A ; 106(1): 138-146, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28875559

RESUMO

Alginate hydrogels have been widely validated for controlled release of growth factors and cytokines, but studies exploring sustained release of small hydrophobic lipids are lacking. Sphingosine-1-phosphate (S1P), a bioactive lipid, is an appealing small molecule for inducing blood vessel formation in the context of ischemic conditions. However, there are numerous biological and engineering challenges associated with designing biomaterial systems for controlled release of this lipid. Thus, the objective of this study was to design an injectable, alginate hydrogel formulation that provides controlled release of S1P to establish locally sustained concentration gradients that promote neovascularization. Herein, we varied the molecular weight distribution of alginate polymers within the hydrogel to alter the resultant mechanical properties in a manner that provides control over S1P release. With increasing high molecular weight (HMW) content, the hydrogels exhibited stiffer material properties and released S1P at slower rates. Accordingly, S1P released from hydrogels with 100% HMW content led to enhanced directed migration of outgrowth endothelial cells and blood vessel development assessed using a chick chorioallantoic membrane assay as compared to hydrogels with less HMW content. Overall, this study describes how alginate hydrogels of varied molecular weight may be used to control S1P release kinetics for therapeutic applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 138-146, 2018.


Assuntos
Alginatos/química , Hidrogéis/química , Lisofosfolipídeos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Esfingosina/análogos & derivados , Materiais Biocompatíveis/química , Movimento Celular/efeitos dos fármacos , Preparações de Ação Retardada/química , Sistemas de Liberação de Medicamentos , Feminino , Sangue Fetal/citologia , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Células Endoteliais da Veia Umbilical Humana , Humanos , Lisofosfolipídeos/química , Peso Molecular , Cultura Primária de Células , Esfingosina/química , Esfingosina/farmacologia
2.
Ann Biomed Eng ; 45(4): 1003-1014, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27904998

RESUMO

Sphingosine-1-phosphate (S1P), a bioactive lipid, is a potent candidate for treatment of ischemic vascular disease. However, designing biomaterial systems for the controlled release of S1P to achieve therapeutic angiogenesis presents both biological and engineering challenges. Thus, the objective of this study was to design a hydrogel system that provides controlled and sustained release of S1P to establish local concentration gradients that promote neovascularization. Alginate hydrogels have been extensively studied and characterized for delivery of proangiogenic factors. We sought to explore if chitosan (0, 0.1, 0.5, or 1%) incorporation could be used as a means to control S1P release from alginate hydrogels. With increasing chitosan incorporation, hydrogels exhibited significantly denser pore structure and stiffer material properties. While 0.1 and 0.5% chitosan gels demonstrated slower respective release of S1P, release from 1% chitosan gels was similar to alginate gels alone. Furthermore, 0.5% chitosan gels induced greater sprouting and directed migration of outgrowth endothelial cells (OECs) in response to released S1P under hypoxia in vitro. Overall, this report presents a platform for a novel alginate-chitosan hydrogel of controlled composition and in situ gelation properties that can be used to control lipid release for therapeutic applications.


Assuntos
Alginatos , Quitosana , Células Endoteliais da Veia Umbilical Humana/metabolismo , Hidrogéis , Lisofosfolipídeos , Neovascularização Fisiológica/efeitos dos fármacos , Esfingosina/análogos & derivados , Alginatos/química , Alginatos/farmacocinética , Alginatos/farmacologia , Células Cultivadas , Quitosana/química , Quitosana/farmacocinética , Quitosana/farmacologia , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Preparações de Ação Retardada/farmacologia , Ácido Glucurônico/química , Ácido Glucurônico/farmacocinética , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/química , Ácidos Hexurônicos/farmacocinética , Ácidos Hexurônicos/farmacologia , Humanos , Hidrogéis/química , Hidrogéis/farmacocinética , Hidrogéis/farmacologia , Lisofosfolipídeos/química , Lisofosfolipídeos/farmacocinética , Lisofosfolipídeos/farmacologia , Esfingosina/química , Esfingosina/farmacocinética , Esfingosina/farmacologia
3.
Plast Reconstr Surg ; 138(6): 1231-1241, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27879591

RESUMO

BACKGROUND: The authors compared the endothelial differentiation capacities of human and rat adipose-derived stem cells to determine whether human adipose-derived stem cells can be a source of endothelial cells clinically. METHODS: Human and rat adipose-derived stem cells were harvested and characterized with flow cytometry and trilineage differentiation. Cells from passages III through V were fed with endothelial cell differentiation medium for up to 3 weeks. Cells were harvested after 1, 2, and 3 weeks, and endothelial differentiation was evaluated with quantitative reverse-transcriptase polymerase chain reaction, flow cytometry, and angiogenic sprouting assays. RESULTS: Both human and rat adipose-derived stem cells were CD90, CD44, and CD31 before differentiation. The cells were successfully differentiated into adipogenic, osteogenic, and chondrogenic lineages. Expression of endothelial cell-specific genes peaked at the second week of differentiation in both human and rat cells. The fold changes in expression of CD31, vascular endothelial growth factor receptor-1, nitric oxide synthase, and von Willebrand factor genes at week 2 were 0.4 ± 0.1, 34.7 ± 0.3, 2.03 ± 0.25, and 12.5 ± 0.3 respectively, in human adipose-derived stem cells; and 1.5 ± 1.01, 21.6 ± 1.7, 17.9 ± 0.6, and 11.2 ± 1.3, respectively, in rat cells. The percentages of CD31 cells were 0.2, 0.64, and 1.6 in human cell populations and 0.5, 5.91, and 11.5 in rat cell populations at weeks 1, 2, and 3, respectively. Rat adipose-derived stem cell-derived endothelial cells displayed enhanced sprouting capability compared with the human cells. CONCLUSIONS: Human adipose-derived stem cells responded less strongly to EGM-2MV endothelial differentiation medium than did the rat cells. Still, the human cells have the potential to become a clinical source of endothelial cells with modifications in the differentiation conditions.


Assuntos
Diferenciação Celular , Células Endoteliais/fisiologia , Células-Tronco Mesenquimais/fisiologia , Gordura Subcutânea/citologia , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Meios de Cultura , Citometria de Fluxo , Humanos , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Control Release ; 237: 42-9, 2016 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-27374631

RESUMO

Hydrogels are an especially appealing class of biomaterials for gene delivery vehicles as they can be introduced into the body with minimally invasive procedures and are often applied in tissue engineering and regenerative medicine strategies. In this study, we show for the first time the use of an injectable alginate hydrogel for controlled delivery of lentivectors in the skeletal muscle of murine hindlimb. We propose to alter the release rates of lentivectors through manipulation of the molecular weight distribution of alginate hydrogels. The release of lentivector was tested using two different ratios of low and high molecular weight (MW) alginate polymers (75/25 and 25/75 low/high MW). The interdependency of lentivector release rate and alginate degradation rate was assessed in vitro. Lentivector-loaded hydrogels maintained transduction potential for up to one week in vitro as demonstrated by the continual transduction of HEK-293T cells. Injection of lentivector-loaded hydrogel in vivo led to a sustained level of transgene expression for more than two months while minimizing the copies of lentivirus genome inserted into the genome of murine skeletal muscle cells. This strategy of spatiotemporal control of lentivector delivery from alginate hydrogels may provide a versatile tool to combine gene therapy and biomaterials for applications in regenerative medicine.


Assuntos
Alginatos/química , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Lentivirus/genética , Músculo Esquelético/metabolismo , Transdução Genética/métodos , Alginatos/administração & dosagem , Animais , Materiais Biocompatíveis/administração & dosagem , Materiais Biocompatíveis/química , Feminino , Vetores Genéticos/genética , Ácido Glucurônico/administração & dosagem , Ácido Glucurônico/química , Células HEK293 , Ácidos Hexurônicos/administração & dosagem , Ácidos Hexurônicos/química , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Injeções , Camundongos , Camundongos Endogâmicos BALB C , Engenharia Tecidual , Transgenes
5.
Ann Biomed Eng ; 43(10): 2301-13, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26206680

RESUMO

Poor vascular homeostasis drives many clinical disorders including diabetes, arthritis, atherosclerosis, and peripheral artery disease. Local tissue ischemia resultant of insufficient blood flow is a potent stimulus for recruitment of endothelial progenitor cells (EPCs). This mobilization and homing is a multi-step process involving EPC detachment from their steady state bone marrow niches, entry into circulation, rolling along vessel endothelium, transmigration, and adhesion to denuded extracellular matrix (ECM) where they may participate in neovessel formation. However, these events are often interrupted in pathological conditions partly due to an imbalance in factor presentation at the tissue level. EPC number and function is impaired in patients with vascular diseases and this dysfunction has been proposed as a prominent contributor to disease pathogenesis. Research approaches aimed at providing therapeutic angiogenesis commonly involve the delivery of proangiogenic cells and/or soluble factors. Nevertheless, greater understanding of the mechanisms involved in EPC homing in both healthy and diseased states is critical for improving efficacy of such strategies. This review underscores the matrix-related signals necessary for enhancing EPC recruitment to ischemic tissue and provides an overview of the development of synthetic ECMs that aim to mimic functions of the local native microenvironment for use in therapeutic angiogenesis.


Assuntos
Células Endoteliais , Matriz Extracelular , Neovascularização Fisiológica , Transplante de Células-Tronco , Células-Tronco , Doenças Vasculares , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/transplante , Mobilização de Células-Tronco Hematopoéticas , Humanos , Isquemia/metabolismo , Isquemia/patologia , Isquemia/terapia , Células-Tronco/metabolismo , Células-Tronco/patologia , Migração Transendotelial e Transepitelial , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia , Doenças Vasculares/terapia
6.
Tissue Eng Part B Rev ; 21(6): 531-42, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26035484

RESUMO

The presentation and controlled release of bioactive signals to direct cellular growth and differentiation represents a widely used strategy in tissue engineering. Historically, work in this field has primarily focused on the delivery of large cytokines and growth factors, which can be costly to manufacture and difficult to deliver in a sustained manner. There has been a marked increase over the past decade in the pursuit of lipid mediators due to their wide range of effects over multiple cell types, low cost, and ease of scale-up. Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are two bioactive lysophospholipids (LPLs) that have gained attention for use as pharmacological agents in tissue engineering applications. While these lipids can have similar effects on cellular response, they possess distinct chemical backbones, mechanisms of synthesis and degradation, and signaling pathways using a discrete set of G-protein-coupled receptors (GPCRs). LPA and S1P predominantly act extracellularly on their GPCRs and can directly regulate cell survival, differentiation, cytokine secretion, proliferation, and migration--each of the important functions that must be considered in regenerative medicine. In addition to these potent physiological functions, these LPLs play pivotal roles in a number of pathophysiological processes. To capitalize on the promise of these molecules in tissue engineering, these lipids have been incorporated into biomaterials for in vivo delivery. Here, we survey the effects of LPA and S1P on both cellular- and tissue-level phenotypes, with an eye toward regulating stem/progenitor cell growth and differentiation. In particular, we examine work that has translational applications for cell-based tissue engineering strategies in promoting cell survival, bone and cartilage engineering, and therapeutic angiogenesis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Esfingosina/análogos & derivados , Células-Tronco/metabolismo , Engenharia Tecidual/métodos , Animais , Movimento Celular/efeitos dos fármacos , Humanos , Lisofosfolipídeos/metabolismo , Esfingosina/metabolismo , Esfingosina/farmacologia
7.
PLoS One ; 10(4): e0123437, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25875493

RESUMO

Therapeutic angiogenesis provides a promising approach to treat ischemic cardiovascular diseases through the delivery of proangiogenic cells and/or molecules. Outgrowth endothelial cells (OECs) are vascular progenitor cells that are especially suited for therapeutic strategies given their ease of noninvasive isolation from umbilical cord or adult peripheral blood and their potent ability to enhance tissue neovascularization. These cells are recruited to sites of vascular injury or tissue ischemia and directly incorporate within native vascular endothelium to participate in neovessel formation. A better understanding of how OEC activity may be boosted under hypoxia with external stimulation by proangiogenic molecules remains a challenge to improving their therapeutic potential. While vascular endothelial growth factor (VEGF) is widely established as a critical factor for initiating angiogenesis, sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, has recently gained great enthusiasm as a potential mediator in neovascularization strategies. This study tests the hypothesis that hypoxia and the presence of VEGF impact the angiogenic response of OECs to S1P stimulation in vitro. We found that hypoxia altered the dynamically regulated S1P receptor 1 (S1PR1) expression on OECs in the presence of S1P (1.0 µM) and/or VEGF (1.3 nM). The combined stimuli of S1P and VEGF together promoted OEC angiogenic activity as assessed by proliferation, wound healing, 3D sprouting, and directed migration under both normoxia and hypoxia. Hypoxia substantially augmented the response to S1P alone, resulting in ~6.5-fold and ~25-fold increases in sprouting and directed migration, respectively. Overall, this report highlights the importance of establishing hypoxic conditions in vitro when studying ischemia-related angiogenic strategies employing vascular progenitor cells.


Assuntos
Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Oxigênio/farmacologia , Esfingosina/análogos & derivados , Células-Tronco/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Alginatos/química , Bioensaio , Hipóxia Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Expressão Gênica/efeitos dos fármacos , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hidrogéis , Neovascularização Fisiológica/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Células-Tronco/citologia , Células-Tronco/metabolismo , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...